Network


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

Hotspot


Dive into the research topics where Aimee S. Payne is active.

Publication


Featured researches published by Aimee S. Payne.


Journal of The American Academy of Dermatology | 2008

Consensus statement on definitions of disease, end points, and therapeutic response for pemphigus

Dédée F. Murrell; Sarah Dick; A. R. Ahmed; Masayuki Amagai; Maria A. Barnadas; Luca Borradori; Jean Claude Bystryn; Giuseppe Cianchini; Luis A. Diaz; David P. Fivenson; Russell P. Hall; K. E. Harman; Takashi Hashimoto; Michael Hertl; Nico Hunzelmann; Pilar Iranzo; Pascal Joly; Marcel F. Jonkman; Yasuo Kitajima; Neil J. Korman; Linda K. Martin; Daniel Mimouni; Amit G. Pandya; Aimee S. Payne; David S. Rubenstein; Hiroshi Shimizu; Animesh Sinha; David Sirois; Detlef Zillikens; Victoria P. Werth

Our scientific knowledge of pemphigus has dramatically progressed in recent years. However, despite the availability of various therapeutic options for the treatment of inflammatory diseases, only a few multicenter controlled trials have helped to define effective therapies in pemphigus. A major obstacle in comparing therapeutic outcomes between centers is the lack of generally accepted definitions and measurements for the clinical evaluation of patients with pemphigus. Common terms and end points of pemphigus are needed so that experts in the field can accurately measure and assess disease extent, activity, severity, and therapeutic response, and thus facilitate and advance clinical trials. This consensus statement from the International Pemphigus Committee represents 2 years of collaborative efforts to attain mutually acceptable common definitions for pemphigus. These should assist in development of consistent reporting of outcomes in future studies.


Journal of Clinical Investigation | 2005

Genetic and functional characterization of human pemphigus vulgaris monoclonal autoantibodies isolated by phage display

Aimee S. Payne; Ken Ishii; Stephen Kacir; Chenyan Lin; Hong Li; Yasushi Hanakawa; Kazuyuki Tsunoda; Masayuki Amagai; John R. Stanley; Don L. Siegel

Pemphigus is a life-threatening blistering disorder of the skin and mucous membranes caused by pathogenic autoantibodies to desmosomal adhesion proteins desmoglein 3 (Dsg3) and Dsg1. Mechanisms of antibody pathogenicity are difficult to characterize using polyclonal patient sera. Using antibody phage display, we have isolated repertoires of human anti-Dsg mAbs as single-chain variable-region fragments (scFvs) from a patient with active mucocutaneous pemphigus vulgaris. ScFv mAbs demonstrated binding to Dsg3 or Dsg1 alone, or both Dsg3 and Dsg1. Inhibition ELISA showed that the epitopes defined by these scFvs are blocked by autoantibodies from multiple pemphigus patients. Injection of scFvs into neonatal mice identified 2 pathogenic scFvs that caused blisters histologically similar to those observed in pemphigus patients. Similarly, these 2 scFvs, but not others, induced cell sheet dissociation of cultured human keratinocytes, indicating that both pathogenic and nonpathogenic antibodies were isolated. Genetic analysis of these mAbs showed restricted patterns of heavy and light chain gene usage, which were distinct for scFvs with different desmoglein-binding specificities. Detailed characterization of these pemphigus mAbs should lead to a better understanding of the immunopathogenesis of disease and to more specifically targeted therapeutic approaches.


Journal of The American Academy of Dermatology | 2012

Definitions and outcome measures for bullous pemphigoid: recommendations by an international panel of experts

Dédée F. Murrell; Benjamin S. Daniel; Pascal Joly; Luca Borradori; Masayuki Amagai; Takashi Hashimoto; F. Caux; Branka Marinović; Animesh A. Sinha; Michael Hertl; Philippe Bernard; David A. Sirois; Giuseppe Cianchini; Janet A. Fairley; Marcel F. Jonkman; Amit G. Pandya; David S. Rubenstein; Detlef Zillikens; Aimee S. Payne; David T. Woodley; Giovanna Zambruno; Valeria Aoki; Carlo Pincelli; Luis A. Diaz; Russell P. Hall; Michael Meurer; José M. Mascaró; Enno Schmidt; Hiroshi Shimizu; John J. Zone

Our scientific knowledge of bullous pemphigoid (BP) has dramatically progressed in recent years. However, despite the availability of various therapeutic options for the treatment of inflammatory diseases, only a few multicenter controlled trials have helped to define effective therapies in BP. A major obstacle in sharing multicenter-based evidences for therapeutic efforts is the lack of generally accepted definitions for the clinical evaluation of patients with BP. Common terms and end points of BP are needed so that experts in the field can accurately measure and assess disease extent, activity, severity, and therapeutic response, and thus facilitate and advance clinical trials. These recommendations from the International Pemphigoid Committee represent 2 years of collaborative efforts to attain mutually acceptable common definitions for BP and proposes a disease extent score, the BP Disease Area Index. These items should assist in the development of consistent reporting of outcomes in future BP reports and studies.


Journal of Investigative Dermatology | 2009

Reliability and Convergent Validity of Two Outcome Instruments for Pemphigus

Misha Rosenbach; Dédée F. Murrell; Jean Claude Bystryn; Sam Dulay; Sarah Dick; Steve Fakharzadeh; Russell P. Hall; Neil J. Korman; Julie Lin; Joyce Okawa; Amit G. Pandya; Aimee S. Payne; Mathew Rose; David S. Rubenstein; David T. Woodley; Carmela C. Vittorio; Benjamin Werth; Erik A. Williams; Lynne Taylor; Andrea B. Troxel; Victoria P. Werth

A major obstacle in performing multicenter controlled trials for pemphigus is the lack of a validated disease activity scoring system. Here, we assess the reliability and convergent validity of the PDAI (pemphigus disease area index). A group of 10 dermatologists scored 15 patients with pemphigus to estimate the inter- and intra-rater reliability of the PDAI and the recently described ABSIS (autoimmune bullous skin disorder intensity score) instrument. To assess convergent validity, these tools were also correlated with the Physicians Global Assessment (PGA). Reliability studies demonstrated an intra-class correlation coefficient (ICC) for inter-rater reliability of 0.76 (95% confirdence interval (CI)=0.61-0.91) for the PDAI and 0.77 (0.63-0.91) for the ABSIS. The tools differed most in reliability of assessing skin activity, with an ICC of 0.39 (0.17-0.60) for the ABSIS and 0.86 (0.76-0.95) for the PDAI. Intra-rater test-retest reliability demonstrated an ICC of 0.98 (0.96-1.0) for the PDAI and 0.80 (0.65-0.96) for the ABSIS. The PDAI also correlated more closely with the PGA. We conclude that the PDAI is more reproducible and correlates better with physician impression of extent. Subset analysis suggests that for this population of mild-to-moderate disease activity, the PDAI captures more variability in cutaneous disease than the ABSIS.


Science | 2016

Reengineering chimeric antigen receptor T cells for targeted therapy of autoimmune disease

Christoph T. Ellebrecht; Vijay Bhoj; Arben Nace; Eun Jung Choi; Xuming Mao; Michael Jeffrey Cho; Giovanni Di Zenzo; Antonio Lanzavecchia; John T. Seykora; George Cotsarelis; Michael C. Milone; Aimee S. Payne

Engineering T cells to treat autoimmunity Autoimmune diseases such as lupus and rheumatoid arthritis lack therapies that specifically target only the disease-causing cells. Inspired by the clinical success of using chimeric antigen receptor T cells to treat certain types of cancers, Ellebrecht et al. asked whether a similar approach might also work against antibody-driven autoimmune diseases. They engineered T cells to express chimeric receptors consisting of the disease-causing autoantigen desmoglein 3 fused to signaling domains that activate T cells. When given to diseased mice, the engineered T cells targeted and killed B cells that express antibodies targeting desmoglein 3, hinting that such a strategy may be an effective way to treat antibody-driven autoimmune diseases. Science, this issue p. 179 A proof-of-principle study indicates that engineered T cells may be an effective, targeted therapy for autoimmunity. Ideally, therapy for autoimmune diseases should eliminate pathogenic autoimmune cells while sparing protective immunity, but feasible strategies for such an approach have been elusive. Here, we show that in the antibody-mediated autoimmune disease pemphigus vulgaris (PV), autoantigen-based chimeric immunoreceptors can direct T cells to kill autoreactive B lymphocytes through the specificity of the B cell receptor (BCR). We engineered human T cells to express a chimeric autoantibody receptor (CAAR), consisting of the PV autoantigen, desmoglein (Dsg) 3, fused to CD137-CD3ζ signaling domains. Dsg3 CAAR-T cells exhibit specific cytotoxicity against cells expressing anti-Dsg3 BCRs in vitro and expand, persist, and specifically eliminate Dsg3-specific B cells in vivo. CAAR-T cells may provide an effective and universal strategy for specific targeting of autoreactive B cells in antibody-mediated autoimmune disease.


Archives of Dermatology | 2012

Adjuvant rituximab therapy of pemphigus: a single center experience with 31 patients

Luisa Lunardon; Kathleen J. Tsai; Kathleen J. Propert; Nicole Fett; John R. Stanley; Victoria P. Werth; Donald E. Tsai; Aimee S. Payne

BACKGROUND We conducted a retrospective study of patients with pemphigus vulgaris (n = 24) and foliaceus (n = 7) treated with adjuvant rituximab to determine efficacy and adverse events. The end point for efficacy was complete remission of disease taking no or minimal therapy. OBSERVATIONS Eighteen patients (58%) achieved the study end point. Of these, 13 patients achieved complete remission off systemic therapy. Patients achieving the study end point had a median disease duration before rituximab therapy of 19 months vs 86 months in those not achieving the end point (P = .01). For the 18 patients achieving the end point, the median (SD) duration of remission was 19 (2) months. Eight of these 18 patients (44%) relapsed from 6 to 17 months after treatment. Serious adverse events attributed to rituximab treatment (osteomyelitis or phlegmon) occurred in 2 patients (6%). In paired serum samples from 10 patients before and after rituximab treatment, the percent change in serum desmoglein index value (median, -80%) was unrelated to the percent change in pneumococcal antibodies (median, +8%) (Spearman rank correlation coefficient r = -0.2). CONCLUSIONS Patients treated with rituximab earlier in the course of disease may have better outcomes. A discussion of rituximabs mechanism of action supports the rationale for early therapy. Prospective clinical studies are necessary to substantiate this observation.


American Journal of Pathology | 2010

Autoimmunity to Desmocollin 3 in Pemphigus Vulgaris

Xuming Mao; Arielle R. Nagler; Sara A. Farber; Eun Jung Choi; Lauren H. Jackson; Kristin M. Leiferman; Norito Ishii; Takashi Hashimoto; Masayuki Amagai; John J. Zone; Aimee S. Payne

Pemphigus vulgaris is a blistering disease associated with autoantibodies to the desmosomal adhesion protein, desmoglein 3. Genetic deficiency of desmoglein 3 in mice mimics autoimmunity to desmoglein 3 in pemphigus vulgaris, with mucosal-dominant blistering in the suprabasal layer of the epidermis. Mice with an epidermal-specific deletion of desmocollin 3, the other major desmosomal cadherin isoform expressed in the basal epidermis, develop suprabasal blisters in skin that are histologically identical to those observed in pemphigus vulgaris, suggesting that desmocollin 3 might be a target of autoantibodies in some pemphigus vulgaris patients. We now demonstrate that desmocollin 3 is an autoantigen in pemphigus vulgaris, illustrated in a patient with mucosal-dominant blistering. Six of 38 pemphigus vulgaris and one of 85 normal serum samples immunoprecipitate desmocollin 3 (P = 0.003). Incubation of patient IgG with human keratinocytes causes loss of intercellular adhesion, and adsorption with recombinant desmocollin 3 specifically prevents this pathogenic effect. Additionally, anti-desmocollin 3 sera cause loss of keratinocyte cell surface desmocollin 3, but not desmoglein 3 by immunofluorescence, indicating distinct cellular pathogenic effects in anti-desmocollin and anti-desmoglein pemphigus, despite their identical clinical presentations. These data demonstrate that desmocollin 3 is a pathogenic autoantigen in pemphigus vulgaris and suggest that pemphigus vulgaris is a histological reaction pattern that may result from autoimmunity to desmoglein 3, desmocollin 3, or both desmosomal cadherins.


Journal of Investigative Dermatology | 2009

Disruption of Desmosome Assembly by Monovalent Human Pemphigus Vulgaris Monoclonal Antibodies

Xuming Mao; Eun Jung Choi; Aimee S. Payne

The intercellular interactions of the desmosomal cadherins, desmoglein and desmocollin, are required for epidermal cell adhesion. Pemphigus vulgaris (PV) is a potentially fatal autoimmune blistering disease characterized by autoantibodies against desmoglein (Dsg) 3. During calcium-induced desmosome assembly, treatment of primary human keratinocytes with pathogenic monovalent anti-Dsg3 mAbs produced from a PV patient causes a decrease of Dsg3 and desmoplakin but not desmocollin (Dsc) 3 in the Triton-insoluble fraction of cell lysates within 2 hours. Immunofluorescence and antibody ELISA studies suggest that pathogenic mAbs cause internalization of cell-surface Dsg3 but not Dsc3 through early endosomes. Electron microscopy demonstrated a lack of well-formed desmosomes in keratinocytes treated with pathogenic compared to nonpathogenic mAbs. In contrast, pathogenic mAbs caused late depletion of Dsg3 from preformed desmosomes at 24 hours, with effects on multiple desmosomal proteins including Dsc3 and plakoglobin. Together, these studies indicate that pathogenic PV mAbs specifically cause internalization of newly synthesized Dsg3 during desmosome assembly, correlating with their pathogenic activity. Monovalent human PV anti-Dsg mAbs reproduce the effects of polyclonal PV IgG on Dsg3 and will facilitate future studies to further dissect the cellular mechanisms for the loss of cell adhesion in pemphigus.


Current Molecular Medicine | 2014

Molecular Diagnosis in Autoimmune Skin Blistering Conditions

J.V. Otten; Takashi Hashimoto; Michael Hertl; Aimee S. Payne; Cassian Sitaru

Blister formation in skin and mucous membranes results from a loss of cell-cell or cell-matrix adhesion and is a common outcome of pathological events in a variety of conditions, including autoimmune and genetic diseases, viral and bacterial infections, or injury by physical and chemical factors. Autoantibodies against structural components maintaining cell-cell and cell-matrix adhesion induce tissue damage in autoimmune blistering diseases. Detection of these autoantibodies either tissue-bound or circulating in serum is essential to diagnose the autoimmune nature of disease. Various immunofluorescence methods as well as molecular immunoassays, including enzyme-linked immunosorbent assay and immunoblotting, belong to the modern diagnostic algorithms for these disorders. There is still a considerable need to increase awareness of the rare autoimmune blistering diseases, which often show a severe, chronic-relapsing course, among physicians and the public. This review article describes the immunopathological features of autoimmune bullous diseases and the molecular immunoassays currently available for their diagnosis and monitoring.


PLOS ONE | 2012

Signaling Dependent and Independent Mechanisms in Pemphigus Vulgaris Blister Formation

Masataka Saito; Sara N. Stahley; Christopher Y. Caughman; Xuming Mao; Dana K. Tucker; Aimee S. Payne; Masayuki Amagai; Andrew P. Kowalczyk

Pemphigus vulgaris (PV) is an autoimmune epidermal blistering disease caused by autoantibodies directed against the desmosomal cadherin desmoglein-3 (Dsg3). Significant advances in our understanding of pemphigus pathomechanisms have been derived from the generation of pathogenic monoclonal Dsg3 antibodies. However, conflicting models for pemphigus pathogenicity have arisen from studies using either polyclonal PV patient IgG or monoclonal Dsg3 antibodies. In the present study, the pathogenic mechanisms of polyclonal PV IgG and monoclonal Dsg3 antibodies were directly compared. Polyclonal PV IgG cause extensive clustering and endocytosis of keratinocyte cell surface Dsg3, whereas pathogenic mouse monoclonal antibodies compromise cell-cell adhesion strength without causing these alterations in Dsg3 trafficking. Furthermore, tyrosine kinase or p38 MAPK inhibition prevents loss of keratinocyte adhesion in response to polyclonal PV IgG. In contrast, disruption of adhesion by pathogenic monoclonal antibodies is not prevented by these inhibitors either in vitro or in human skin explants. Our results reveal that the pathogenic activity of polyclonal PV IgG can be attributed to p38 MAPK-dependent clustering and endocytosis of Dsg3, whereas pathogenic monoclonal Dsg3 antibodies can function independently of this pathway. These findings have important implications for understanding pemphigus pathophysiology, and for the design of pemphigus model systems and therapeutic interventions.

Collaboration


Dive into the Aimee S. Payne's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xuming Mao

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

John R. Stanley

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Eun Jung Choi

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Victoria P. Werth

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Don L. Siegel

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eric M. Mukherjee

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Michael C. Milone

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge