Network


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

Hotspot


Dive into the research topics where Hannah J. Barbian is active.

Publication


Featured researches published by Hannah J. Barbian.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Phenotypic properties of transmitted founder HIV-1

Nicholas F. Parrish; Feng Gao; Hui Li; Elena E. Giorgi; Hannah J. Barbian; Erica H. Parrish; Lara Zajic; Shilpa S. Iyer; Julie M. Decker; Amit Kumar; Bhavna Hora; Anna Berg; Fangping Cai; Jennifer Hopper; Thomas N. Denny; Hairao Ding; Christina Ochsenbauer; John C. Kappes; Rachel P. Galimidi; Anthony P. West; Pamela J. Bjorkman; Craig B. Wilen; Robert W. Doms; Meagan O'Brien; Nina Bhardwaj; Persephone Borrow; Barton F. Haynes; Mark Muldoon; James Theiler; Bette T. Korber

Defining the virus–host interactions responsible for HIV-1 transmission, including the phenotypic requirements of viruses capable of establishing de novo infections, could be important for AIDS vaccine development. Previous analyses have failed to identify phenotypic properties other than chemokine receptor 5 (CCR5) and CD4+ T-cell tropism that are preferentially associated with viral transmission. However, most of these studies were limited to examining envelope (Env) function in the context of pseudoviruses. Here, we generated infectious molecular clones of transmitted founder (TF; n = 27) and chronic control (CC; n = 14) viruses of subtypes B (n = 18) and C (n = 23) and compared their phenotypic properties in assays specifically designed to probe the earliest stages of HIV-1 infection. We found that TF virions were 1.7-fold more infectious (P = 0.049) and contained 1.9-fold more Env per particle (P = 0.048) compared with CC viruses. TF viruses were also captured by monocyte-derived dendritic cells 1.7-fold more efficiently (P = 0.035) and more readily transferred to CD4+ T cells (P = 0.025). In primary CD4+ T cells, TF and CC viruses replicated with comparable kinetics; however, when propagated in the presence of IFN-α, TF viruses replicated to higher titers than CC viruses. This difference was significant for subtype B (P = 0.000013) but not subtype C (P = 0.53) viruses, possibly reflecting demographic differences of the respective patient cohorts. Together, these data indicate that TF viruses are enriched for higher Env content, enhanced cell-free infectivity, improved dendritic cell interaction, and relative IFN-α resistance. These viral properties, which likely act in concert, should be considered in the development and testing of AIDS vaccines.


Journal of Virology | 2012

Mucosal Simian Immunodeficiency Virus Transmission in African Green Monkeys: Susceptibility to Infection Is Proportional to Target Cell Availability at Mucosal Sites

Ivona Pandrea; Nicholas F. Parrish; Kevin Raehtz; Thaidra Gaufin; Hannah J. Barbian; Dongzhu Ma; Jan Kristoff; Rajeev Gautam; Fang Zhong; George Haret-Richter; Anita Trichel; George M. Shaw; Beatrice H. Hahn; Cristian Apetrei

ABSTRACT African green monkeys (AGMs) are naturally infected with a simian immunodeficiency virus (SIVagm) that is nonpathogenic in its host. Although SIVagm is common and widespread, little is known about the mechanisms that govern its transmission. Since the earliest virus-host interactions may provide key insights into the nonpathogenic phenotype of SIVagm, we developed a mucosal transmission model for this virus. Using plasma from an acutely infected AGM as the virus inoculum, we exposed adult and juvenile AGMs, as well as pigtailed macaques (PTMs) as a nonnatural host control, by mucosal routes to increasing titers of virus and compared the doses needed to establish a productive infection. Four juvenile and four adult AGMs as well as two PTMs were intrarectally (IR) exposed, while two additional adult female AGMs were intravaginally (IVAG) exposed. No animal became infected following exposure to 105 RNA copies. Both PTMs but none of the AGMs became infected following exposure to 106 RNA copies. Finally, all adult AGMs and two of the four juvenile AGMs became infected following exposure to 107 RNA copies, acquiring either one (2 IR infected juveniles, 1 IR infected adult, 2 IVAG infected adults) or two (3 IR infected adults) transmitted founder viruses. These results were consistent with immunophenotypic data, which revealed a significant correlation between the percentage of CD4+ T cells expressing CCR5 in the mucosa and the susceptibility to infection, in terms of both the viral dose and the numbers of transmitted founder viruses. Moreover, studies of uninfected AGMs showed that the fraction of CCR5-expressing CD4+ T cells increased significantly with age. These results indicate that (i) AGMs are readily infected with SIVagm by both intrarectal and intravaginal routes, (ii) susceptibility to infection is proportional to the number of available CCR5+ CD4+ target cells in the mucosa, and (iii) the paucity of CCR5+ CD4+ target cells in infant and juvenile AGMs may explain the near absence of vertical transmission.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Resistance to type 1 interferons is a major determinant of HIV-1 transmission fitness

Shilpa S. Iyer; Frederic Bibollet-Ruche; Scott Sherrill-Mix; Gerald H. Learn; Lindsey J. Plenderleith; Andrew G. Smith; Hannah J. Barbian; Ronnie M. Russell; Marcos V. P. Gondim; Catherine Y. Bahari; Christiana M. Shaw; Yingying Li; Timothy Decker; Barton F. Haynes; George M. Shaw; Paul M. Sharp; Persephone Borrow; Beatrice H. Hahn

Significance Effective prevention strategies are urgently needed to control the spread of HIV-1. A critical barrier to developing such strategies is the lack of understanding of the host antiviral defenses that control HIV-1 replication in the mucosa at the site of entry. Here, we characterized viruses from matched donor and recipient pairs to determine whether transmitted HIV-1 strains exhibit traits that increase their transmission fitness. Characterizing 300 limiting dilution-derived isolates, we identified several properties that enhance virus replication in the face of a vigorous innate immune response, of which resistance to type 1 IFNs is the most important. These results provide new insights into the HIV-1 transmission process and define possible new targets for AIDS prevention and therapy. Sexual transmission of HIV-1 is an inefficient process, with only one or few variants of the donor quasispecies establishing the new infection. A critical, and as yet unresolved, question is whether the mucosal bottleneck selects for viruses with increased transmission fitness. Here, we characterized 300 limiting dilution-derived virus isolates from the plasma, and in some instances genital secretions, of eight HIV-1 donor and recipient pairs. Although there were no differences in the amount of virion-associated envelope glycoprotein, recipient isolates were on average threefold more infectious (P = 0.0001), replicated to 1.4-fold higher titers (P = 0.004), were released from infected cells 4.2-fold more efficiently (P < 0.00001), and were significantly more resistant to type I IFNs than the corresponding donor isolates. Remarkably, transmitted viruses exhibited 7.8-fold higher IFNα2 (P < 0.00001) and 39-fold higher IFNβ (P < 0.00001) half-maximal inhibitory concentrations (IC50) than did donor isolates, and their odds of replicating in CD4+ T cells at the highest IFNα2 and IFNβ doses were 35-fold (P < 0.00001) and 250-fold (P < 0.00001) greater, respectively. Interestingly, pretreatment of CD4+ T cells with IFNβ, but not IFNα2, selected donor plasma isolates that exhibited a transmitted virus-like phenotype, and such viruses were also detected in the donor genital tract. These data indicate that transmitted viruses are phenotypically distinct, and that increased IFN resistance represents their most distinguishing property. Thus, the mucosal bottleneck selects for viruses that are able to replicate and spread efficiently in the face of a potent innate immune response.


Journal of Virology | 2013

Heterogeneity in Neutralization Sensitivities of Viruses Comprising the Simian Immunodeficiency Virus SIVsmE660 Isolate and Vaccine Challenge Stock

Michael Lopker; Juliet L. Easlick; Sarah Sterrett; Julie M. Decker; Hannah J. Barbian; Gerald H. Learn; Brandon F. Keele; James E. Robinson; Hui Li; Beatrice H. Hahn; George M. Shaw; Katharine J. Bar

ABSTRACT The sooty mangabey-derived simian immunodeficiency virus (SIV) strain E660 (SIVsmE660) is a genetically heterogeneous, pathogenic isolate that is commonly used as a vaccine challenge strain in the nonhuman primate (NHP) model of human immunodeficiency virus type 1 (HIV-1) infection. Though it is often employed to assess antibody-based vaccine strategies, its sensitivity to antibody-mediated neutralization has not been well characterized. Here, we utilize single-genome sequencing and infectivity assays to analyze the neutralization sensitivity of the uncloned SIVsmE660 isolate, individual viruses comprising the isolate, and transmitted/founder (T/F) viruses arising from low-dose mucosal inoculation of macaques with the isolate. We found that the SIVsmE660 isolate overall was highly sensitive to neutralization by SIV-infected macaque plasma samples (50% inhibitory concentration [IC50] < 10−5) and monoclonal antibodies targeting V3 (IC50 < 0.01 μg/ml), CD4-induced (IC50 < 0.1 μg/ml), CD4 binding site (IC50 ∼ 1 μg/ml), and V4 (IC50, ∼5 μg/ml) epitopes. In comparison, SIVmac251 and SIVmac239 were highly resistant to neutralization by these same antibodies. Differences in neutralization sensitivity between SIVsmE660 and SIVmac251/239 were not dependent on the cell type in which virus was produced or tested. These findings indicate that in comparison to SIVmac251/239 and primary HIV-1 viruses, SIVsmE660 generally exhibits substantially less masking of antigenically conserved Env epitopes. Interestingly, we identified a minor population of viruses (∼10%) in both the SIVsmE660 isolate and T/F viruses arising from it that were substantially more resistant (>1,000-fold) to antibody neutralization and another fraction (∼20%) that was intermediate in neutralization resistance. These findings may explain the variable natural history and variable protection afforded by heterologous Env-based vaccines in rhesus macaques challenged by high-dose versus low-dose SIVsmE660 inoculation regimens.


Mbio | 2015

Neutralization Properties of Simian Immunodeficiency Viruses Infecting Chimpanzees and Gorillas

Hannah J. Barbian; Julie M. Decker; Frederic Bibollet-Ruche; Rachel P. Galimidi; Anthony P. West; Gerald H. Learn; Nicholas F. Parrish; Shilpa S. Iyer; Yingying Li; Craig S. Pace; Ruijiang Song; Yaoxing Huang; Thomas N. Denny; Hugo Mouquet; Loïc Martin; Priyamvada Acharya; Baoshan Zhang; Peter D. Kwong; John R. Mascola; C. Theo Verrips; Nika M. Strokappe; Lucy Rutten; Laura E. McCoy; Robin A. Weiss; Corrine S. Brown; Raven Jackson; Guido Silvestri; Mark Connors; Dennis R. Burton; George M. Shaw

ABSTRACT Broadly cross-reactive neutralizing antibodies (bNabs) represent powerful tools to combat human immunodeficiency virus type 1 (HIV-1) infection. Here, we examined whether HIV-1-specific bNabs are capable of cross-neutralizing distantly related simian immunodeficiency viruses (SIVs) infecting central (Pan troglodytes troglodytes) (SIVcpzPtt) and eastern (Pan troglodytes schweinfurthii) (SIVcpzPts) chimpanzees (n = 11) as well as western gorillas (Gorilla gorilla gorilla) (SIVgor) (n = 1). We found that bNabs directed against the CD4 binding site (n = 10), peptidoglycans at the base of variable loop 3 (V3) (n = 5), and epitopes at the interface of surface (gp120) and membrane-bound (gp41) envelope glycoproteins (n = 5) failed to neutralize SIVcpz and SIVgor strains. In addition, apex V2-directed bNabs (n = 3) as well as llama-derived (heavy chain only) antibodies (n = 6) recognizing both the CD4 binding site and gp41 epitopes were either completely inactive or neutralized only a fraction of SIVcpzPtt strains. In contrast, one antibody targeting the membrane-proximal external region (MPER) of gp41 (10E8), functional CD4 and CCR5 receptor mimetics (eCD4-Ig, eCD4-Igmim2, CD4-218.3-E51, and CD4-218.3-E51-mim2), as well as mono- and bispecific anti-human CD4 (iMab and LM52) and CCR5 (PRO140, PRO140-10E8) receptor antibodies neutralized >90% of SIVcpz and SIVgor strains with low-nanomolar (0.13 to 8.4 nM) potency. Importantly, the latter antibodies blocked virus entry not only in TZM-bl cells but also in Cf2Th cells expressing chimpanzee CD4 and CCR5 and neutralized SIVcpz in chimpanzee CD4+ T cells, with 50% inhibitory concentrations (IC50s) ranging from 3.6 to 40.5 nM. These findings provide new insight into the protective capacity of anti-HIV-1 bNabs and identify candidates for further development to combat SIVcpz infection. IMPORTANCE SIVcpz is widespread in wild-living chimpanzees and can cause AIDS-like immunopathology and clinical disease. HIV-1 infection of humans can be controlled by antiretroviral therapy; however, treatment of wild-living African apes with current drug regimens is not feasible. Nonetheless, it may be possible to curb the spread of SIVcpz in select ape communities using vectored immunoprophylaxis and/or therapy. Here, we show that antibodies and antibody-like inhibitors developed to combat HIV-1 infection in humans are capable of neutralizing genetically diverse SIVcpz and SIVgor strains with considerable breadth and potency, including in primary chimpanzee CD4+ T cells. These reagents provide an important first step toward translating intervention strategies currently developed to treat and prevent AIDS in humans to SIV-infected apes. SIVcpz is widespread in wild-living chimpanzees and can cause AIDS-like immunopathology and clinical disease. HIV-1 infection of humans can be controlled by antiretroviral therapy; however, treatment of wild-living African apes with current drug regimens is not feasible. Nonetheless, it may be possible to curb the spread of SIVcpz in select ape communities using vectored immunoprophylaxis and/or therapy. Here, we show that antibodies and antibody-like inhibitors developed to combat HIV-1 infection in humans are capable of neutralizing genetically diverse SIVcpz and SIVgor strains with considerable breadth and potency, including in primary chimpanzee CD4+ T cells. These reagents provide an important first step toward translating intervention strategies currently developed to treat and prevent AIDS in humans to SIV-infected apes.


American Journal of Primatology | 2018

Destabilization of the gut microbiome marks the end-stage of simian immunodeficiency virus infection in wild chimpanzees

Hannah J. Barbian; Yingying Li; Miguel Ángel Ramírez; Zachary Klase; Iddi Lipende; Deus Mjungu; Andrew H. Moeller; Michael L. Wilson; Anne E. Pusey; Elizabeth V. Lonsdorf; Frederic D. Bushman; Beatrice H. Hahn

Enteric dysbiosis is a characteristic feature of progressive human immunodeficiency virus type 1 (HIV‐1) infection but has not been observed in simian immunodeficiency virus (SIVmac)‐infected macaques, including in animals with end‐stage disease. This has raised questions concerning the mechanisms underlying the HIV‐1 associated enteropathy, with factors other than virus infection, such as lifestyle and antibiotic use, implicated as playing possible causal roles. Simian immunodeficiency virus of chimpanzees (SIVcpz) is also associated with increased mortality in wild‐living communities, and like HIV‐1 and SIVmac, can cause CD4+ T cell depletion and immunodeficiency in infected individuals. Given the central role of the intestinal microbiome in mammalian health, we asked whether gut microbial constituents could be identified that are indicative of SIVcpz status and/or disease progression. Here, we characterized the gut microbiome of SIVcpz‐infected and ‐uninfected chimpanzees in Gombe National Park, Tanzania. Subjecting a small number of fecal samples (N = 9) to metagenomic (shotgun) sequencing, we found bacteria of the family Prevotellaceae to be enriched in SIVcpz‐infected chimpanzees. However, 16S rRNA gene sequencing of a larger number of samples (N = 123) failed to show significant differences in both the composition and diversity (alpha and beta) of gut bacterial communities between infected (N = 24) and uninfected (N = 26) chimpanzees. Similarly, chimpanzee stool‐associated circular virus (Chi‐SCV) and chimpanzee adenovirus (ChAdV) identified by metagenomic sequencing were neither more prevalent nor more abundant in SIVcpz‐infected individuals. However, fecal samples collected from SIVcpz‐infected chimpanzees within 5 months before their AIDS‐related death exhibited significant compositional changes in their gut bacteriome. These data indicate that SIVcpz‐infected chimpanzees retain a stable gut microbiome throughout much of their natural infection course, with a significant destabilization of bacterial (but not viral) communities observed only in individuals with known immunodeficiency within the last several months before their death. Am. J. Primatol. 80:e22515, 2018.


American Journal of Primatology | 2018

Socioecological correlates of clinical signs in two communities of wild chimpanzees (Pan troglodytes) at Gombe National Park, Tanzania

Elizabeth V. Lonsdorf; Thomas R. Gillespie; Tiffany M. Wolf; Iddi Lipende; Jane Raphael; Jared S. Bakuza; Carson M. Murray; Michael L. Wilson; Shadrack Kamenya; Deus Mjungu; D. Anthony Collins; Ian C. Gilby; Margaret A. Stanton; Karen A. Terio; Hannah J. Barbian; Yingying Li; Miguel Ángel Ramírez; Alexander Krupnick; Emily Seidl; Jane Goodall; Beatrice H. Hahn; Anne E. Pusey; Dominic A. Travis

Disease and other health hazards pose serious threats to the persistence of wild ape populations. The total chimpanzee population at Gombe National Park, Tanzania, has declined from an estimated 120 to 150 individuals in the 1960s to around 100 individuals by the end of 2013, with death associated with observable signs of disease as the leading cause of mortality. In 2004, we began a non‐invasive health‐monitoring program in the two habituated communities in the park (Kasekela and Mitumba) with the aim of understanding the prevalence of health issues in the population, and identifying the presence and impacts of various pathogens. Here we present prospectively collected data on clinical signs (observable changes in health) in the chimpanzees of the Kasekela (n = 81) and Mitumba (n = 32) communities over an 8‐year period (2005–2012). First, we take a population approach and analyze prevalence of clinical signs in five different categories: gastrointestinal system (diarrhea), body condition (estimated weight loss), respiratory system (coughing, sneezing etc.), wounds/lameness, and dermatologic issues by year, month, and community membership. Mean monthly prevalence of each clinical sign per community varied, but typically affected <10% of observed individuals. Secondly, we analyze the presence of clinical signs in these categories as they relate to individual demographic and social factors (age, sex, and dominance rank) and simian immunodeficiency virus (SIVcpz) infection status. Adults have higher odds of being observed with diarrhea, loss of body condition, and wounds or lameness when compared to immatures, while males have a higher probability of being observed with wounds or lameness than females. In contrast, signs of respiratory illness appear not to be related to chimpanzee‐specific factors and skin abnormalities are very rare. For a subset of known‐rank individuals, dominance rank predicts the probability of wounding/lameness in adult males, but does not predict any adverse clinical signs in adult females. Instead, adult females with SIVcpz infection are more likely to be observed with diarrhea, a finding that warrants further investigation. Comparable data are needed from other sites to determine whether the prevalence of clinical signs we observe are relatively high or low, as well as to more fully understand the factors influencing health of wild apes at both the population and individual level. Am. J. Primatol. 80:e22562, 2018.


Ecology and Evolution | 2018

CHIIMP: An automated high-throughput microsatellite genotyping platform reveals greater allelic diversity in wild chimpanzees

Hannah J. Barbian; Andrew Connell; Alexa N. Avitto; Ronnie M. Russell; Andrew G. Smith; Madhurima S. Gundlapally; Alexander L. Shazad; Yingying Li; Frederic Bibollet-Ruche; Emily E. Wroblewski; Deus Mjungu; Elizabeth V. Lonsdorf; Fiona A. Stewart; Alex K. Piel; Anne E. Pusey; Paul M. Sharp; Beatrice H. Hahn

Abstract Short tandem repeats (STRs), also known as microsatellites, are commonly used to noninvasively genotype wild‐living endangered species, including African apes. Until recently, capillary electrophoresis has been the method of choice to determine the length of polymorphic STR loci. However, this technique is labor intensive, difficult to compare across platforms, and notoriously imprecise. Here we developed a MiSeq‐based approach and tested its performance using previously genotyped fecal samples from long‐term studied chimpanzees in Gombe National Park, Tanzania. Using data from eight microsatellite loci as a reference, we designed a bioinformatics platform that converts raw MiSeq reads into locus‐specific files and automatically calls alleles after filtering stutter sequences and other PCR artifacts. Applying this method to the entire Gombe population, we confirmed previously reported genotypes, but also identified 31 new alleles that had been missed due to sequence differences and size homoplasy. The new genotypes, which increased the allelic diversity and heterozygosity in Gombe by 61% and 8%, respectively, were validated by replicate amplification and pedigree analyses. This demonstrated inheritance and resolved one case of an ambiguous paternity. Using both singleplex and multiplex locus amplification, we also genotyped fecal samples from chimpanzees in the Greater Mahale Ecosystem in Tanzania, demonstrating the utility of the MiSeq‐based approach for genotyping nonhabituated populations and performing comparative analyses across field sites. The new automated high‐throughput analysis platform (available at https://github.com/ShawHahnLab/chiimp) will allow biologists to more accurately and effectively determine wildlife population size and structure, and thus obtain information critical for conservation efforts.


Cell Reports | 2018

Completeness of HIV-1 Envelope Glycan Shield at Transmission Determines Neutralization Breadth

Kshitij Wagh; Edward F. Kreider; Yingying Li; Hannah J. Barbian; Gerald H. Learn; Elena E. Giorgi; Peter Hraber; Timothy Decker; Andrew G. Smith; Marcos V. P. Gondim; Lindsey Gillis; Jamie Wandzilak; Gwo-Yu Chuang; Reda Rawi; Fangping Cai; Pierre Pellegrino; Ian Williams; Julie Overbaugh; Feng Gao; Peter D. Kwong; Barton F. Haynes; George M. Shaw; Persephone Borrow; Michael S. Seaman; Beatrice H. Hahn; Bette T. Korber

SUMMARY Densely arranged N-linked glycans shield the HIV-1 envelope (Env) trimer from antibody recognition. Strain-specific breaches in this shield (glycan holes) can be targets of vaccine-induced neutralizing antibodies that lack breadth. To understand the interplay between glycan holes and neutralization breadth in HIV-1 infection, we developed a sequence-and structure-based approach to identify glycan holes for individual Env sequences that are shielded in most M-group viruses. Applying this approach to 12 longitudinally followed individuals, we found that transmitted viruses with more intact glycan shields correlated with development of greater neutralization breadth. Within 2 years, glycan acquisition filled most glycan holes present at transmission, indicating escape from hole-targeting neutralizing antibodies. Glycan hole filling generally preceded the time to first detectable breadth, although time intervals varied across hosts. Thus, completely glycan-shielded viruses were associated with accelerated neutralization breadth development, suggesting that Env immunogens with intact glycan shields may be preferred components of AIDS vaccines.


Retrovirology | 2017

Effective treatment of SIVcpz-induced immunodeficiency in a captive western chimpanzee

Hannah J. Barbian; Raven Jackson-Jewett; Corrine S. Brown; Frederic Bibollet-Ruche; Gerald H. Learn; Timothy Decker; Edward F. Kreider; Yingying Li; Thomas N. Denny; Paul M. Sharp; George M. Shaw; Jeffrey D. Lifson; Edward P. Acosta; Michael S. Saag; Katharine J. Bar; Beatrice H. Hahn

Collaboration


Dive into the Hannah J. Barbian's collaboration.

Top Co-Authors

Avatar

Beatrice H. Hahn

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Yingying Li

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

George M. Shaw

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Gerald H. Learn

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Andrew G. Smith

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Deus Mjungu

Jane Goodall Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge