Network


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

Hotspot


Dive into the research topics where Julie A. Karl is active.

Publication


Featured researches published by Julie A. Karl.


Diabetes | 2010

Mesenchymal Stem Cells Enhance Allogeneic Islet Engraftment in Nonhuman Primates

Dora M. Berman; Melissa Willman; Dongmei Han; Gary Kleiner; Norman M. Kenyon; Over Cabrera; Julie A. Karl; Roger W. Wiseman; David H. O'Connor; Amelia Bartholomew; Norma S. Kenyon

OBJECTIVE To test the graft-promoting effects of mesenchymal stem cells (MSCs) in a cynomolgus monkey model of islet/bone marrow transplantation. RESEARCH DESIGN AND METHODS Cynomolgus MSCs were obtained from iliac crest aspirate and characterized through passage 11 for phenotype, gene expression, differentiation potential, and karyotype. Allogeneic donor MSCs were cotransplanted intraportally with islets on postoperative day (POD) 0 and intravenously with donor marrow on PODs 5 and 11. Recipients were followed for stabilization of blood glucose levels, reduction of exogenous insulin requirement (EIR), C-peptide levels, changes in peripheral blood T regulatory cells, and chimerism. Destabilization of glycemia and increases in EIR were used as signs of rejection; additional intravenous MSCs were administered to test the effect on reversal of rejection. RESULTS MSC phenotype and a normal karyotype were observed through passage 11. IL-6, IL-10, vascular endothelial growth factor, TGF-β, hepatocyte growth factor, and galectin-1 gene expression levels varied among donors. MSC treatment significantly enhanced islet engraftment and function at 1 month posttransplant (n = 8), as compared with animals that received islets without MSCs (n = 3). Additional infusions of donor or third-party MSCs resulted in reversal of rejection episodes and prolongation of islet function in two animals. Stable islet allograft function was associated with increased numbers of regulatory T-cells in peripheral blood. CONCLUSIONS MSCs may provide an important approach for enhancement of islet engraftment, thereby decreasing the numbers of islets needed to achieve insulin independence. Furthermore, MSCs may serve as a new, safe, and effective antirejection therapy.


Nature Medicine | 2009

Major histocompatibility complex genotyping with massively parallel pyrosequencing

Roger W. Wiseman; Julie A. Karl; Benjamin N. Bimber; Claire O'Leary; Simon M. Lank; Jennifer J. Tuscher; Ann M. Detmer; Pascal Bouffard; Natalya Levenkova; Cynthia L Turcotte; Edward Szekeres; Chris L. Wright; Timothy T. Harkins; David H. O'Connor

Major histocompatibility complex (MHC) genetics dictate adaptive cellular immune responses, making robust MHC genotyping methods essential for studies of infectious disease, vaccine development and transplantation. Nonhuman primates provide essential preclinical models for these areas of biomedical research. Unfortunately, given the unparalleled complexity of macaque MHCs, existing methodologies are inadequate for MHC typing of these key model animals. Here we use pyrosequencing of complementary DNA–PCR amplicons as a general approach to determine comprehensive MHC class I genotypes in nonhuman primates. More than 500 unique MHC class I sequences were resolved by sequence-based typing of rhesus, cynomolgus and pig-tailed macaques, nearly half of which have not been reported previously. The remarkable sensitivity of this approach in macaques demonstrates that pyrosequencing is viable for ultra–high-throughput MHC genotyping of primates, including humans.


Immunogenetics | 2007

Comprehensive characterization of MHC class II haplotypes in Mauritian cynomolgus macaques

Shelby L. O’Connor; Alex J. Blasky; Chad J. Pendley; Ericka A. Becker; Roger W. Wiseman; Julie A. Karl; Austin L. Hughes; David H. O’Connor

There are currently no nonhuman primate models with fully defined major histocompatibility complex (MHC) class II genetics. We recently showed that six common MHC haplotypes account for essentially all MHC diversity in cynomolgus macaques (Macaca fascicularis) from the island of Mauritius. In this study, we employ complementary DNA cloning and sequencing to comprehensively characterize full length MHC class II alleles expressed at the Mafa-DPA, -DPB, -DQA, -DQB, -DRA, and -DRB loci on the six common haplotypes. We describe 34 full-length MHC class II alleles, 12 of which are completely novel. Polymorphism was evident at all six loci including DPA, a locus thought to be monomorphic in rhesus macaques. Similar to other Old World monkeys, Mauritian cynomolgus macaques (MCM) share MHC class II allelic lineages with humans at the DQ and DR loci, but not at the DP loci. Additionally, we identified extensive sharing of MHC class II alleles between MCM and other nonhuman primates. The characterization of these full-length-expressed MHC class II alleles will enable researchers to generate MHC class II transferent cell lines, tetramers, and other molecular reagents that can be used to explore CD4+ T lymphocyte responses in MCM.


Immunogenetics | 2008

Identification of MHC class I sequences in Chinese-origin rhesus macaques

Julie A. Karl; Roger W. Wiseman; Kevin J. Campbell; Alex J. Blasky; Austin L. Hughes; Betsy Ferguson; Daniel S. Read; David H. O’Connor

The rhesus macaque (Macaca mulatta) is an excellent model for human disease and vaccine research. Two populations exhibiting distinctive morphological and physiological characteristics, Indian- and Chinese-origin rhesus macaques, are commonly used in research. Genetic analysis has focused on the Indian macaque population, but the accessibility of these animals for research is limited. Due to their greater availability, Chinese rhesus macaques are now being used more frequently, particularly in vaccine and biodefense studies, although relatively little is known about their immunogenetics. In this study, we discovered major histocompatibility complex (MHC) class I cDNAs in 12 Chinese rhesus macaques and detected 41 distinct Mamu-A and Mamu-B sequences. Twenty-seven of these class I cDNAs were novel, while six and eight of these sequences were previously reported in Chinese and Indian rhesus macaques, respectively. We then performed microsatellite analysis on DNA from these 12 animals, as well as an additional 18 animals, and developed sequence specific primer PCR (PCR-SSP) assays for eight cDNAs found in multiple animals. We also examined our cohort for potential admixture of Chinese and Indian origin animals using a recently developed panel of single nucleotide polymorphisms (SNPs). The discovery of 27 novel MHC class I sequences in this analysis underscores the genetic diversity of Chinese rhesus macaques and contributes reagents that will be valuable for studying cellular immunology in this population.


Immunogenetics | 2010

Characterization of Mauritian cynomolgus macaque major histocompatibility complex class I haplotypes by high-resolution pyrosequencing

Melisa L. Budde; Roger W. Wiseman; Julie A. Karl; Bozena Hanczaruk; Birgitte B. Simen; David H. O’Connor

Major histocompatibility complex (MHC) class I alleles of nonhuman primates have been associated with disease susceptibility, resistance, and resolution. Here, using high-resolution pyrosequencing, we characterized MHC class I transcripts expressed in Mauritian cynomolgus macaques (MCM), a nonhuman primate population with restricted MHC diversity. Using this approach, we identified 67 distinct MHC class I transcripts encoded by the seven most frequent MCM MHC class I haplotypes, 40 (60%) of which span the complete open reading frames. These results double the number of MHC class I sequences previously defined by cloning and Sanger sequencing of cDNA-PCR products and provide a rapid, high-throughput, and economical method for MHC characterization. Overall, this approach significantly expanded our knowledge of MCM haplotypes and will facilitate future studies on disease pathogenesis and protective cellular immunity.


Journal of Virology | 2009

Mauritian Cynomolgus Macaques Share Two Exceptionally Common Major Histocompatibility Complex Class I Alleles That Restrict Simian Immunodeficiency Virus-Specific CD8+ T Cells

Benjamin J. Burwitz; Chad J. Pendley; Justin M. Greene; Ann M. Detmer; Jennifer J. Lhost; Julie A. Karl; Shari M. Piaskowski; Richard Rudersdorf; Lyle T. Wallace; Benjamin N. Bimber; John T. Loffredo; Daryl G. Cox; Wilfried Bardet; William H. Hildebrand; Roger W. Wiseman; Shelby L. O'Connor; David H. O'Connor

ABSTRACT Vaccines that elicit CD8+ T-cell responses are routinely tested for immunogenicity in nonhuman primates before advancement to clinical trials. Unfortunately, the magnitude and specificity of vaccine-elicited T-cell responses are variable in currently utilized nonhuman primate populations, owing to heterogeneity in major histocompatibility (MHC) class I genetics. We recently showed that Mauritian cynomolgus macaques (MCM) have unusually simple MHC genetics, with three common haplotypes encoding a shared pair of MHC class IA alleles, Mafa-A*25 and Mafa-A*29. Based on haplotype frequency, we hypothesized that CD8+ T-cell responses restricted by these MHC class I alleles would be detected in nearly all MCM. We examine here the frequency and functionality of these two alleles, showing that 88% of MCM express Mafa-A*25 and Mafa-A*29 and that animals carrying these alleles mount three newly defined simian immunodeficiency virus-specific CD8+ T-cell responses. The epitopes recognized by each of these responses accumulated substitutions consistent with immunologic escape, suggesting these responses exert antiviral selective pressure. The demonstration that Mafa-A*25 and Mafa-A*29 restrict CD8+ T-cell responses that are shared among nearly all MCM indicates that these animals are an advantageous nonhuman primate model for comparing the immunogenicity of vaccines that elicit CD8+ T-cell responses.


Science Translational Medicine | 2010

MHC Heterozygote Advantage in Simian Immunodeficiency Virus–Infected Mauritian Cynomolgus Macaques

Shelby L. O'Connor; Jennifer J. Lhost; Ericka A. Becker; Ann M. Detmer; Randall C. Johnson; Caitlin E. MacNair; Roger W. Wiseman; Julie A. Karl; Justin M. Greene; Benjamin J. Burwitz; Benjamin N. Bimber; Simon M. Lank; Jennifer J. Tuscher; Edward T. Mee; Nicola J. Rose; Ronald C. Desrosiers; Austin L. Hughes; Thomas C. Friedrich; Mary Carrington; David H. O'Connor

This manuscript demonstrates unambiguous major histocompatibility complex heterozygote advantage in macaque monkeys infected with the same strain of simian immunodeficiency virus, suggesting that a prophylactic HIV vaccine should elicit a population of CD8+ T cells with broad specificity. A Broad View of HIV Some studies of HIV-infected people have suggested that HIV is better controlled when the individual’s immune response is broader, that is, when more parts of the HIV virus are recognized by T cells. Indeed, the lack of a broad immune response may explain why HIV vaccines have generally not been successful. Despite the importance of this question for vaccine design, it has been difficult to answer definitively because of diversity in HIV strain, sampling time after infection, individual genetics, and other variables. Now, O’Connor et al. use genetically defined Mauritian cynomolgus macaques to get around these issues and test whether a broader immune response does in fact lead to better disease control. The immune response to a virus is determined in part by the genetics at the HLA locus. This locus is important because variability in HLA class I genes determines the number of major histocompatibility complex (MHC) molecules generated; the number of MHC molecules then determines the number of epitopes that can be presented to immune CD8 T cells. Individuals who are heterozygotes at this locus are expected to have a broader immune response than do homozygotes because they have the potential to present a more diverse set of epitopes to immune cells. O’Connor and colleagues measured viral blood concentrations and cellular immune responses in cynomolgus macaques harboring identical MHC genetics and infected with the same strain of simian immunodeficiency virus; this enabled them to unambiguously define the relationship among MHC diversity, CD8 T cell breadth, and disease outcome. They found that the vast majority of macaques homozygous for MHC had viral loads nearly 80 times those of their heterozygote counterparts; the associated CD8 T cell responses, measured by immune assays that rely on visualization techniques, were inconsistent. Therefore, to better understand their results, the authors examined how the animals’ CD8 T cell epitopes changed with time. They found that viral sequences isolated from MHC heterozygotes collected 1 year after infection matched variants observed in each of their MHC homozygote counterparts at 1 year after infection, which suggested that the CD8 T cell responses in MHC heterozygotes were an assemblage of the responses from their MHC homozygote counterparts. These data collectively indicate that the potential breadth of the immune response determines viral replication: The broader the response, the less replication. This study builds on previous observational studies showing heterozygote advantage in HIV-infected people, and sets the stage for future studies exploring the mechanisms responsible for this immunological control of immunodeficiency viruses. Furthermore, through the use of these macaques with identical MHC genetics, vaccine candidates can be tested for their effectiveness in the presence of limited CD8 T lymphocyte diversity. The importance of a broad CD8 T lymphocyte (CD8-TL) immune response to HIV is unknown. Ex vivo measurements of immunological activity directed at a limited number of defined epitopes provide an incomplete portrait of the actual immune response. We examined viral loads in simian immunodeficiency virus (SIV)–infected major histocompatibility complex (MHC)–homozygous and MHC-heterozygous Mauritian cynomolgus macaques. Chronic viremia in MHC-homozygous macaques was 80 times that in MHC-heterozygous macaques. Virus from MHC-homozygous macaques accumulated 11 to 14 variants, consistent with escape from CD8-TL responses after 1 year of SIV infection. The pattern of mutations detected in MHC-heterozygous macaques suggests that their epitope-specific CD8-TL responses are a composite of those present in their MHC-homozygous counterparts. These results provide the clearest example of MHC heterozygote advantage among individuals infected with the same immunodeficiency virus strain, suggesting that broad recognition of multiple CD8-TL epitopes should be a key feature of HIV vaccines.


American Journal of Transplantation | 2010

Phenotype, Distribution and Alloreactive Properties of Memory T Cells from Cynomolgus Monkeys

O. Nadazdin; S. Boskovic; Toru Murakami; David H. O'Connor; Roger W. Wiseman; Julie A. Karl; Jennifer J. Tuscher; David H. Sachs; Joren C. Madsen; Georges Tocco; Tatsuo Kawai; Cosimi Ab; Gilles Benichou

The high frequency of memory T cells present in primates is thought to represent a major barrier to tolerance induction in transplantation. Therefore, it is crucial to characterize these memory T cells and determine their functional properties. High numbers of memory T cells were detected in peripheral blood and all lymphoid tissues except lymph nodes, which were essentially the site of naïve T cells. The majority of CD8+ memory T cells were effector memory cells located in the blood and bone marrow while most CD4+ memory T cells were central memory cells present in the spleen. Next, memory T cells from over 100 monkeys were tested for their response to alloantigens by ELISPOT. Memory alloreactivity mediated via direct but not indirect allorecognition was detected in all animals. The frequency of allospecific memory T cells varied dramatically depending upon the nature of the responder/stimulator monkey combination tested. MHC gene matching was generally associated with a low‐memory alloreactivity. Nevertheless, low anamnestic alloresponses were also found in a significant number of fully MHC‐mismatched monkey combinations. These results show that selected donor/recipient combinations displaying a low memory alloresponsiveness can be found. These combinations may be more favorable for transplant tolerance induction.


Journal of Medical Primatology | 2009

MHC haplotype frequencies in a UK breeding colony of Mauritian cynomolgus macaques mirror those found in a distinct population from the same geographic origin.

Edward T. Mee; Anjna Badhan; Julie A. Karl; Roger W. Wiseman; Keith L Cutler; Leslie A. Knapp; Neil Almond; David H. O’Connor; Nicola J. Rose

Background  Mauritian cynomolgus macaques have greatly restricted genetic diversity in the MHC region compared to other non‐human primates; however, the frequency of common MHC haplotypes among captive‐bred populations has not been reported.


Ilar Journal | 2013

Haplessly Hoping: Macaque Major Histocompatibility Complex Made Easy

Roger W. Wiseman; Julie A. Karl; Patrick S. Bohn; Francesca A. Nimityongskul; Gabriel J. Starrett; David H. O'Connor

Major histocompatibility complex (MHC) gene products control the repertoire of T cell responses that an individual may create against pathogens and foreign tissues. This text will review the current understanding of MHC genetics in nonhuman primates, with a focus on Mauritian-origin cynomolgus macaques (Macaca fascicularis) and Indian-origin rhesus macaques (Macaca mulatta). These closely related macaque species provide important experimental models for studies of infectious disease pathogenesis, vaccine development, and transplantation research. Recent advances resulting from the application of several cost effective, high-throughput approaches, with deep sequencing technologies have revolutionized our ability to perform MHC genotyping of large macaque cohorts. Pyrosequencing of cDNA amplicons with a Roche/454 GS Junior instrument, provides excellent resolution of MHC class I allelic variants with semi-quantitative estimates of relative levels of transcript abundance. Introduction of the Illumina MiSeq platform significantly increased the sample throughput, since the sample loading workflow is considerably less labor intensive, and each instrument run yields approximately 100-fold more sequence data. Extension of these sequencing methods from cDNA to genomic DNA amplicons further streamlines the experimental workflow and opened opportunities for retrospective MHC genotyping of banked DNA samples. To facilitate the reporting of MHC genotypes, and comparisons between groups of macaques, this text also introduces an intuitive series of abbreviated rhesus MHC haplotype designations based on a major Mamu-A or Mamu-B transcript characteristic for ancestral allele combinations. The authors believe that the use of MHC-defined macaques promises to improve the reproducibility, and predictability of results from pre-clinical studies for translation to humans.

Collaboration


Dive into the Julie A. Karl's collaboration.

Top Co-Authors

Avatar

Roger W. Wiseman

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

David H. O’Connor

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

David H. O'Connor

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Benjamin N. Bimber

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Simon M. Lank

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Austin L. Hughes

University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Ericka A. Becker

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Alex J. Blasky

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Jennifer J. Tuscher

University of Wisconsin-Madison

View shared research outputs
Top Co-Authors

Avatar

Patrick S. Bohn

University of Wisconsin-Madison

View shared research outputs
Researchain Logo
Decentralizing Knowledge