Network


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

Hotspot


Dive into the research topics where Natalie A. Hutnick is active.

Publication


Featured researches published by Natalie A. Hutnick.


Science | 2012

Innate Lymphoid Cells Promote Anatomical Containment of Lymphoid-Resident Commensal Bacteria

Gregory F. Sonnenberg; Laurel A. Monticelli; Theresa Alenghat; Thomas C. Fung; Natalie A. Hutnick; Jun Kunisawa; Naoko Shibata; Stephanie Grunberg; Rohini Sinha; Adam M. Zahm; Mélanie R. Tardif; Taheri Sathaliyawala; Masaru Kubota; Donna L. Farber; Ronald G. Collman; Abraham Shaked; Lynette A. Fouser; David B. Weiner; Philippe A. Tessier; Joshua R. Friedman; Hiroshi Kiyono; Frederic D. Bushman; Kyong-Mi Chang; David Artis

Protecting Against a Barrier Breach In order to coexist peacefully, a “firewall” exists that keeps the commensal bacteria that reside in our intestines and associated lymphoid tissue contained. Several diseases and infections, however, lead to a breach in this barrier, which leads to chronic inflammation and pathology. Sonnenberg et al. (p. 1321) found that in mice, innate lymphoid cells (ILCs) are critically important for the anatomical containment of commensal bacteria in an interleukin-22 (IL-22)–dependent manner. ILC depletion or blockade of IL-22 led to loss of bacterial containment and systemic inflammation. Lymphocytes prevent bacteria from spreading beyond gut-associated lymphoid tissues and causing systemic inflammation. The mammalian intestinal tract is colonized by trillions of beneficial commensal bacteria that are anatomically restricted to specific niches. However, the mechanisms that regulate anatomical containment remain unclear. Here, we show that interleukin-22 (IL-22)–producing innate lymphoid cells (ILCs) are present in intestinal tissues of healthy mammals. Depletion of ILCs resulted in peripheral dissemination of commensal bacteria and systemic inflammation, which was prevented by administration of IL-22. Disseminating bacteria were identified as Alcaligenes species originating from host lymphoid tissues. Alcaligenes was sufficient to promote systemic inflammation after ILC depletion in mice, and Alcaligenes-specific systemic immune responses were associated with Crohn’s disease and progressive hepatitis C virus infection in patients. Collectively, these data indicate that ILCs regulate selective containment of lymphoid-resident bacteria to prevent systemic inflammation associated with chronic diseases.


Clinical Infectious Diseases | 2011

Clinical Applications of DNA Vaccines: Current Progress

Bernadette Ferraro; Matthew P. Morrow; Natalie A. Hutnick; Thomas Shin; Colleen E. Lucke; David B. Weiner

It was discovered almost 20 years ago that plasmid DNA, when injected into the skin or muscle of mice, could induce immune responses to encoded antigens. Since that time, there has since been much progress in understanding the basic biology behind this deceptively simple vaccine platform and much technological advancement to enhance immune potency. Among these advancements are improved formulations and improved physical methods of delivery, which increase the uptake of vaccine plasmids by cells; optimization of vaccine vectors and encoded antigens; and the development of novel formulations and adjuvants to augment and direct the host immune response. The ability of the current, or second-generation, DNA vaccines to induce more-potent cellular and humoral responses opens up this platform to be examined in both preventative and therapeutic arenas. This review focuses on these advances and discusses both preventive and immunotherapeutic clinical applications.


Blood | 2009

AAV-1-mediated gene transfer to skeletal muscle in humans results in dose-dependent activation of capsid-specific T cells.

Federico Mingozzi; Janneke J. Meulenberg; Daniel J. Hui; Etiena Basner-Tschakarjan; Nicole C. Hasbrouck; Shyrie Edmonson; Natalie A. Hutnick; Michael R. Betts; John J. P. Kastelein; Erik S.G. Stroes; Katherine A. High

In a clinical trial for adeno-associated virus serotype 1 (AAV-1)-mediated gene transfer to muscle for lipoprotein lipase (LPL) deficiency, 1 subject from the high-dose cohort experienced a transient increase in the muscle enzyme creatine phosphokinase (CPK) 4 weeks after gene transfer. Simultaneously, after an initial downward trend consistent with expression of LPL, plasma triglyceride levels returned to baseline. We characterized B- and T-cell responses to the vector and the transgene product in the subjects enrolled in this study. IFN-gamma enzyme-linked immunosorbent spot (ELISpot) and intracellular cytokine staining assays performed on peripheral blood mononuclear cells (PBMCs) from the subject who experienced the CPK elevation showed the activation of capsid-specific CD4(+) and CD8(+) T cells. Four of 8 subjects had detectable T-cell responses to capsid with dose-dependent kinetics of appearance. Subjects with detectable T-cell responses to capsid also had higher anti-AAV-1 IgG3 antibody titer. No subject developed B- or T-cell responses to the LPL transgene product. These findings suggest that T-cell responses directed to the AAV-1 capsid are dose-dependent. Whether they also limit the duration of expression of the transgene at higher doses is unclear, and will require additional analyses at later time points.


Nature Medicine | 2009

Adenovirus-specific immunity after immunization with an Ad5 HIV-1 vaccine candidate in humans

Kara L. O'Brien; Jinyan Liu; Sharon L. King; Ying-Hua Sun; Joern E. Schmitz; Michelle A. Lifton; Natalie A. Hutnick; Michael R. Betts; Sheri A. Dubey; Jaap Goudsmit; John W. Shiver; Michael N. Robertson; Danilo R. Casimiro; Dan H. Barouch

The immunologic basis for the potential enhanced HIV-1 acquisition in adenovirus serotype 5 (Ad5)-seropositive individuals who received the Merck recombinant Ad5 HIV-1 vaccine in the STEP study remains unclear. Here we show that baseline Ad5-specific neutralizing antibodies are not correlated with Ad5-specific T lymphocyte responses and that Ad5-seropositive subjects do not develop higher vector-specific cellular immune responses as compared with Ad5-seronegative subjects after vaccination. These findings challenge the hypothesis that activated Ad5-specific T lymphocytes were the cause of the potential enhanced HIV-1 susceptibility in the STEP study.


PLOS Pathogens | 2010

Perforin and IL-2 Upregulation Define Qualitative Differences among Highly Functional Virus-Specific Human CD8(+) T Cells

George Makedonas; Natalie A. Hutnick; Danielle Haney; Alexandra C. Amick; Jay Gardner; Gabriela L. Cosma; Adam R. Hersperger; Douglas V. Dolfi; E. John Wherry; Guido Ferrari; Michael R. Betts

The prevailing paradigm of T lymphocyte control of viral replication is that the protective capacity of virus-specific CD8+ T cells is directly proportional to the number of functions they can perform, with IL-2 production capacity considered critical. Having recently defined rapid perforin upregulation as a novel effector function of antigen-specific CD8+ T cells, here we sought to determine whether new perforin production is a component of polyfunctional CD8+ T cell responses that contributes to the control of several human viral infections: cytomegalovirus (CMV), Epstein-Barr virus (EBV), influenza (flu), and adenovirus (Ad). We stimulated normal human donor PBMC with synthetic peptides whose amino acid sequences correspond to defined CTL epitopes in the aforementioned viruses, and then used polychromatic flow cytometry to measure the functional capacity and the phenotype of the responding CD8+ T cells. While EBV and flu-specific CD8+ T cells rarely upregulate perforin, CMV-specific cells often do and Ad stimulates an exceptionally strong perforin response. The differential propensity of CD8+ T cells to produce either IL-2 or perforin is in part related to levels of CD28 and the transcription factor T-bet, as CD8+ T cells that rapidly upregulate perforin harbor high levels of T-bet and those producing IL-2 express high amounts of CD28. Thus, “polyfunctional” profiling of antigen-specific CD8+ T cells must not be limited to simply the number of functions the cell can perform, or one particular memory phenotype, but should actually define which combinations of memory markers and functions are relevant in each pathogenic context.


Nature Medicine | 2009

Baseline Ad5 serostatus does not predict Ad5 HIV vaccine-induced expansion of adenovirus-specific CD4+ T cells.

Natalie A. Hutnick; Diane G. Carnathan; Sheri A. Dubey; George Makedonas; Kara S. Cox; Lisa S. Kierstead; Sarah J. Ratcliffe; Michael N. Robertson; Danilo R. Casimiro; Hildegund C.J. Ertl; Michael R. Betts

The mechanisms underlying possible increased HIV-1 acquisition in adenovirus 5 (Ad5)-seropositive subjects vaccinated with Ad5–HIV-1 vectors in the Merck STEP trial remain unclear. We find that Ad5 serostatus does not predict Ad5-specific CD4+ T cell frequency, and we did not observe durable significant differences in Ad5-specific CD4+ T cells between Ad5-seropositive and Ad5-seronegative subjects after vaccination. These findings indicate no causative role for Ad5-specific CD4+ T cells in increasing HIV-1 susceptibility in the STEP trial.


Vaccine | 2010

Adenovirus-specific human T cells are pervasive, polyfunctional, and cross-reactive.

Natalie A. Hutnick; Diane G. Carnathan; Korey Demers; George Makedonas; Hildegund C.J. Ertl; Michael R. Betts

Pre-existing immunity to adenovirus (Ad) reduces the efficacy of Ad-based vaccines. The goal of this study was to define the prevalence, magnitude, functionality and phenotype of Ad-specific human T cells directly ex vivo. To study the magnitude of T-cell responses to Ad, we developed a highly reproducible whole Ad vector stimulation assay for use with polychromatic flow cytometry. Ad-specific CD4(+) and CD8(+) T-cells were detected in all 17 human subjects tested and were capable of proliferating upon restimulation. Ad5-specific CD4(+) T cells were primarily monofunctional CD4(+) T cells that produced IL-2, IFN-gamma or TNFalpha and expressed the memory markers CD27 and CD45RO. In contrast, Ad5-specific CD8(+) T cells were more polyfunctional, expressing effector-like combinations of IFN-gamma, MIP1alpha and perforin, and generally lacked CD27 and CD45RO expression. Ad-specific CD4(+) and CD8(+) T-cell responses against chimpanzee-derived AdC6 and AdC7 were found in all subjects, indicating the commonality of cross-serotype reactivity of Ad-specific T cells. This cross-reactivity is due in part to extensive CD4(+) and CD8(+) T-cell recognition of hexon regions conserved between multiple Ad serotypes. The prevalence, cross-reactivity and effector-like functions of Ad-specific T cells in humans may affect the efficacy of Ad vector-based vaccines by eliminating vector infected cells even when rare serotype Ad vectors are employed.


Current Opinion in Virology | 2011

Selected approaches for increasing HIV DNA vaccine immunogenicity in vivo.

Natalie A. Hutnick; Dj Myles; Chaoran Billie Bian; Karuppiah Muthumani; David B. Weiner

The safety, stability, and ability for repeat homologous vaccination makes the DNA vaccine platform an excellent candidate for an effective HIV-1 vaccine. However, the immunogenicity of early DNA vaccines did not translate from small animal models into larger non-human primates and was markedly lower than viral vectors. In addition to improvements to the DNA vector itself, delivery with electroporation, the inclusion of molecular adjuvants, and heterologous prime-boost strategies have dramatically improved the immunogenicity of DNA vaccines for HIV and currently makes them a leading platform with many areas warranting further research and clinical development.


Infection and Immunity | 2013

Inducing Humoral and Cellular Responses to Multiple Sporozoite and Liver-Stage Malaria Antigens Using Exogenous Plasmid DNA

Bernadette Ferraro; Kendra T. Talbott; A. Balakrishnan; Neil J. Cisper; Matthew P. Morrow; Natalie A. Hutnick; Dj Myles; Devon J. Shedlock; N Obeng-Adjei; Jian Yan; A. K. K. Kayatani; N. Richie; W. Cabrera; R. Shiver; Amir S. Khan; A. S. Brown; Maria Yang; U. Wille-Reece; A. J. Birkett; Niranjan Y. Sardesai; David B. Weiner

ABSTRACT A vaccine candidate that elicits humoral and cellular responses to multiple sporozoite and liver-stage antigens may be able to confer protection against Plasmodium falciparum malaria; however, a technology for formulating and delivering such a vaccine has remained elusive. Here, we report the preclinical assessment of an optimized DNA vaccine approach that targets four P. falciparum antigens: circumsporozoite protein (CSP), liver stage antigen 1 (LSA1), thrombospondin-related anonymous protein (TRAP), and cell-traversal protein for ookinetes and sporozoites (CelTOS). Synthetic DNA sequences were designed for each antigen with modifications to improve expression and were delivered using in vivo electroporation (EP). Immunogenicity was evaluated in mice and nonhuman primates (NHPs) and assessed by enzyme-linked immunosorbent assay (ELISA), gamma interferon (IFN-γ) enzyme-linked immunosorbent spot (ELISpot) assay, and flow cytometry. In mice, DNA with EP delivery induced antigen-specific IFN-γ production, as measured by ELISpot assay and IgG seroconversion against all antigens. Sustained production of IFN-γ, interleukin-2, and tumor necrosis factor alpha was elicited in both the CD4+ and CD8+ T cell compartments. Furthermore, hepatic CD8+ lymphocytes produced LSA1-specific IFN-γ. The immune responses conferred to mice by this approach translated to the NHP model, which showed cellular responses by ELISpot assay and intracellular cytokine staining. Notably, antigen-specific CD8+ granzyme B+ T cells were observed in NHPs. Collectively, the data demonstrate that delivery of gene sequences by DNA/EP encoding malaria parasite antigens is immunogenic in animal models and can harness both the humoral and cellular arms of the immune system.


PLOS ONE | 2013

HIV-1 Env DNA vaccine plus protein boost delivered by EP expands B- and T-cell responses and neutralizing phenotype in vivo.

Kar Muthumani; Megan C. Wise; Kate E. Broderick; Natalie A. Hutnick; Jonathan Goodman; Seleeke Flingai; Jian Yan; Chaoran B. Bian; Janess Mendoza; Colleen Tingey; Christine Wilson; Krzysztof Wojtak; Niranjan Y. Sardesai; David B. Weiner

An effective HIV vaccine will most likely require the induction of strong T-cell responses, broadly neutralizing antibodies (bNAbs), and the elicitation of antibody-dependent cellular cytotoxicity (ADCC). Previously, we demonstrated the induction of strong HIV/SIV cellular immune responses in macaques and humans using synthetic consensus DNA immunogens delivered via adaptive electroporation (EP). However, the ability of this improved DNA approach to prime for relevant antibody responses has not been previously studied. Here, we investigate the immunogenicity of consensus DNA constructs encoding gp140 sequences from HIV-1 subtypes A, B, C and D in a DNA prime-protein boost vaccine regimen. Mice and guinea pigs were primed with single- and multi-clade DNA via EP and boosted with recombinant gp120 protein. Sera were analyzed for gp120 binding and induction of neutralizing antibody activity. Immunization with recombinant Env protein alone induced low-titer binding antibodies with limited neutralization breath. In contrast, the synthetic DNA prime-protein boost protocol induced significantly higher antibody binding titers. Furthermore, sera from DNA prime-protein boost groups were able to neutralize a broader range of viruses in a panel of tier 1 clade B viruses as well as multiple tier 1 clade A and clade C viruses. Further investigation of synthetic DNA prime plus adaptive EP plus protein boost appears warranted.

Collaboration


Dive into the Natalie A. Hutnick's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jian Yan

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Dj Myles

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Michael R. Betts

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amir S. Khan

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge