Network


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

Hotspot


Dive into the research topics where Laurence B. Peterson is active.

Publication


Featured researches published by Laurence B. Peterson.


Nature Genetics | 2007

Interleukin-2 gene variation impairs regulatory T cell function and causes autoimmunity

Jun Yamanouchi; Dan Rainbow; Pau Serra; Sarah Howlett; Kara Hunter; Valerie Garner; Andrea Gonzalez-Munoz; Jan Clark; Riitta Veijola; Rose M. Cubbon; Show-Ling Chen; Ray Rosa; Anne Marie Cumiskey; David V. Serreze; Simon G. Gregory; Jane Rogers; Paul A. Lyons; Barry Healy; Luc J. Smink; John A. Todd; Laurence B. Peterson; Linda S. Wicker; Pere Santamaria

Autoimmune diseases are thought to result from imbalances in normal immune physiology and regulation. Here, we show that autoimmune disease susceptibility and resistance alleles on mouse chromosome 3 (Idd3) correlate with differential expression of the key immunoregulatory cytokine interleukin-2 (IL-2). In order to test directly that an approximately twofold reduction in IL-2 underpins the Idd3-linked destabilization of immune homeostasis, we show that engineered haplodeficiency of Il2 gene expression not only reduces T cell IL-2 production by twofold but also mimics the autoimmune dysregulatory effects of the naturally occurring susceptibility alleles of Il2. Reduced IL-2 production achieved by either genetic mechanism correlates with reduced function of CD4+ CD25+ regulatory T cells, which are critical for maintaining immune homeostasis.


Diabetes | 1994

β2-Microglobulin–Deficient NOD Mice Do Not Develop Insulitis or Diabetes

Linda S. Wicker; Edward H. Leiter; John A. Todd; Robert J Renjilian; Erin Peterson; Paul Fischer; Patricia L Podolin; Maarten Zijlstra; Rudolf Jaenisch; Laurence B. Peterson

The role of CD8+ T-cells in the development of diabetes in the nonobese diabetic (NOD) mouse remains controversial. Although it is widely agreed that class II-restricted CD4+ T-cells are essential for the development of diabetes in the NOD model, some studies have suggested that CD8+ T-cells are not required for β-cell destruction. To assess the contribution of CD8+ T-cells to diabetes, we have developed a class of NOD mouse that lacks expression of βxs2-microglobulin (NOD-B2mnull). NOD-B2mnull mice, which lack both class I expression and CD8+ T-cells in the periphery, not only failed to develop diabetes but were completely devoid of insulitis. These results demonstrate an essential role for CD8+ T-cells in the initiation of the autoimmune response to β-cells in the NOD mouse.


Immunity | 2004

An autoimmune disease-associated CTLA-4 splice variant lacking the B7 binding domain signals negatively in T cells.

Lalitha Vijayakrishnan; Jacqueline M. Slavik; Zsolt Illes; Rebecca J. Greenwald; Dan Rainbow; Bernhard Greve; Laurence B. Peterson; David A. Hafler; Gordon J. Freeman; Arlene H. Sharpe; Linda S. Wicker; Vijay K. Kuchroo

Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) plays a critical role in downregulating T cell responses. A number of autoimmune diseases have shown genetic linkage to the CTLA-4 locus. We have cloned and expressed an alternatively spliced form of CTLA-4 that has genetic linkage with type I diabetes in the NOD mice. This splice variant of CTLA-4, named ligand-independent CTLA-4 (liCTLA-4), lacks exon2 including the MYPPPY motif essential for binding to the costimulatory ligands B7-1 and B7-2. Here we show that liCTLA-4 is expressed as a protein in primary T cells and strongly inhibits T cell responses by binding and dephosphorylating the TcRzeta chain. Expression of liCTLA-4, but not full-length CTLA-4 (flCTLA-4), was higher in memory/regulatory T cells from diabetes-resistant NOD congenic mice compared to susceptible NOD mice. These data suggest that increased expression and negative signaling delivered by the liCTLA-4 may regulate development of T cell-mediated autoimmune diseases.


Immunity | 2000

The NOD Idd9 Genetic Interval Influences the Pathogenicity of Insulitis and Contains Molecular Variants of Cd30, Tnfr2, and Cd137

Paul A. Lyons; Wayne W. Hancock; Paul W. Denny; Christopher J. Lord; Natasha J Hill; Nicola Armitage; Thorsten Siegmund; John A. Todd; Michael S. Phillips; J. Fred Hess; Shiow-Ling Chen; Paul Fischer; Laurence B. Peterson; Linda S. Wicker

Previous analyses of NOD mice have shown that some genes control the development of both insulitis and diabetes, while other loci influence diabetes without reducing insulitis. Evidence for the existence of a gene only influencing diabetes, Idd9 on mouse chromosome 4, is provided here by the development of a novel congenic mouse strain, NOD.B10 Idd9. NOD.B10 Idd9 mice display profound resistance to diabetes even though nearly all develop insulitis. Subcongenic analysis has demonstrated that alleles of at least three B10 genes, Idd9.1, Idd9.2, and Idd9.3 are required to produce Idd9-mediated diabetes resistance. Candidate genes with amino acid differences between the NOD and B10 strains have been localized to the 5.6 cM Idd9.2 interval (Tnfr2, Cd30) and to the 2.0 cM Idd9.3 interval (Cd137).


Journal of Lipid Research | 2011

A PCSK9-binding antibody that structurally mimics the EGF(A) domain of LDL-receptor reduces LDL cholesterol in vivo

Yan G. Ni; Di Marco S; Jon H. Condra; Laurence B. Peterson; Weirong Wang; Fubao Wang; Shilpa Pandit; Holly A. Hammond; Ray Rosa; Cummings Rt; Dana D Wood; Xiaomei Liu; Bottomley Mj; Xun Shen; Cubbon Rm; Wang Sp; Douglas G. Johns; Volpari C; Hamuro L; Jayne Chin; Lingyi Huang; Jing Zhang Zhao; Salvatore Vitelli; Peter Haytko; Douglas Wisniewski; Lyndon J. Mitnaul; Carl P. Sparrow; Brian K. Hubbard; Andrea Carfi; Ayesha Sitlani

Proprotein convertase subtilisin-like/kexin type 9 (PCSK9) regulates LDL cholesterol levels by inhibiting LDL receptor (LDLr)-mediated cellular LDL uptake. We have identified a fragment antigen-binding (Fab) 1D05 which binds PCSK9 with nanomolar affinity. The fully human antibody 1D05-IgG2 completely blocks the inhibitory effects of wild-type PCSK9 and two gain-of-function human PCSK9 mutants, S127R and D374Y. The crystal structure of 1D05-Fab bound to PCSK9 reveals that 1D05-Fab binds to an epitope on the PCSK9 catalytic domain which includes the entire LDLr EGF(A) binding site. Notably, the 1D05-Fab CDR-H3 and CDR-H2 loops structurally mimic the EGF(A) domain of LDLr. In a transgenic mouse model (CETP/LDLr-hemi), in which plasma lipid and PCSK9 profiles are comparable to those of humans, 1D05-IgG2 reduces plasma LDL cholesterol to 40% and raises hepatic LDLr protein levels approximately fivefold. Similarly, in healthy rhesus monkeys, 1D05-IgG2 effectively reduced LDL cholesterol 20%–50% for over 2 weeks, despite its relatively short terminal half-life (t1/2 = 3.2 days). Importantly, the decrease in circulating LDL cholesterol corresponds closely to the reduction in free PCSK9 levels. Together these results clearly demonstrate that the LDL-lowering effect of the neutralizing anti-PCSK9 1D05-IgG2 antibody is mediated by reducing the amount of PCSK9 that can bind to the LDLr.


Journal of Experimental Medicine | 2006

NOD.c3c4 congenic mice develop autoimmune biliary disease that serologically and pathogenetically models human primary biliary cirrhosis

Junichiro Irie; Yuehong Wu; Linda S. Wicker; Daniel B. Rainbow; Michael A. Nalesnik; Raphael Hirsch; Laurence B. Peterson; Patrick S.C. Leung; Chunmei Cheng; Ian R. Mackay; M. Eric Gershwin; William M. Ridgway

Primary biliary cirrhosis (PBC) is an autoimmune disease with a strong genetic component characterized by biliary ductular inflammation with eventual liver cirrhosis. The serologic hallmark of PBC is antimitochondrial antibodies that react with the pyruvate dehydrogenase complex, targeting the inner lipoyl domain of the E2 subunit (anti–PDC-E2). Herein we demonstrate that NOD.c3c4 mice congenically derived from the nonobese diabetic strain develop an autoimmune biliary disease (ABD) that models human PBC. NOD.c3c4 (at 9–10 wk, before significant biliary pathology) develop antibodies to PDC-E2 that are specific for the inner lipoyl domain. Affected areas of biliary epithelium are infiltrated with CD3+, CD4+, and CD8+ T cells, and treatment of NOD.c3c4 mice with monoclonal antibody to CD3 protects from ABD. Furthermore, NOD.c3c4-scid mice develop disease after adoptive transfer of splenocytes or CD4+ T cells, demonstrating a central role for T cells in pathogenesis. Histological analysis reveals destructive cholangitis, granuloma formation, and eosinophilic infiltration as seen in PBC, although, unlike PBC, the extrahepatic biliary ducts are also affected. Using a congenic mapping approach, we define the first ABD (Abd) locus, Abd1. These results identify the NOD.c3c4 mouse as the first spontaneous mouse model of PBC.


Journal of Clinical Investigation | 1996

Naturally processed T cell epitopes from human glutamic acid decarboxylase identified using mice transgenic for the type 1 diabetes-associated human MHC class II allele, DRB1*0401.

Linda S. Wicker; Shiow Ling Chen; Gerald T. Nepom; John F. Elliott; Daniel C. Freed; Alka Bansal; Song Zheng; Andrew Herman; Åke Lernmark; Dennis M. Zaller; Laurence B. Peterson; Jonathan B. Rothbard; Richard D. Cummings; Phyllis Jonas Whiteley

The identification of class II binding peptide epitopes from autoimmune disease-related antigens is an essential step in the development of antigen-specific immune modulation therapy. In the case of type 1 diabetes, T cell and B cell reactivity to the autoantigen glutamic acid decarboxylase 65 (GAD65) is associated with disease development in humans and in nonobese diabetic (NOD) mice. In this study, we identify two DRB1*0401-restricted T cell epitopes from human GAD65, 274-286, and 115-127. Both peptides are immunogenic in transgenic mice expressing functional DRB1*0401 MHC class II molecules but not in nontransgenic littermates. Processing of GAD65 by antigen presenting cells (APC) resulted in the formation of DRB1*0401 complexes loaded with either the 274-286 or 115-127 epitopes, suggesting that these naturally derived epitopes may be displayed on APC recruited into pancreatic islets. The presentation of these two T cell epitopes in the islets of DRB1*0401 individuals who are at risk for type 1 diabetes may allow for antigen-specific recruitment of regulatory cells to the islets following peptide immunization.


Diabetes | 1995

Crosses of NOD Mice With the Related NON Strain: A Polygenic Model for IDDM

Marcia A. McAleer; Peter C. Reifsnyder; Sheila M. Palmer; Michal Prochazka; Jennifer M. Love; James B. Copeman; Elizabeth E. Powell; Nanda Rodrigues; Jan-Bas Prins; David V. Serreze; Nicole H. DeLarato; Linda S. Wicker; Laurence B. Peterson; Nicholas J. Schork; John A. Todd; Edward H. Leiter

Chromosome locations of non-major histocompatibility complex (MHC) genes contributing to insulin-dependent diabetes mellitus (IDDM) in mice have been determined by outcrossing NOD mice to other inbred strains congenic for the NOD MHC haplotype (H2g7). At least nine non-MHC IDDM susceptibility genes (Idd) were previously identified at first backcross (BC1) after outcross of NOD to C57BL/10.H2g7 congenic mice (B10.H2g7). We investigated whether the same set of Idd loci segregated with IDDM susceptibility after outcross of NOD to NON.H2g7 congenic mice. Since the outcrosses to NON.H2g7 and B10.H2g7 were performed in the same vivarium, direct comparisons were made of the chromosomal locations and relative strengths of Idd alleles in diabetic progeny from the two different outcrosses. In comparison with the NOD x B10.H2g7 outcross, the NOD x NON.H2g7 outcross produced significantly higher IDDM frequencies in F1, F2, and BC1 generations. The high F2 diabetes frequency allowed evaluation of the effects of homozygous expression of both the susceptibility and the resistance allele at Idd loci. This analysis demonstrated that no single non-MHC Idd locus was essential for the onset of diabetes in this cross. After outcross to NON.H2g7, Idd4 (chromosome [Chr] 11), Idd5 (Chr 1), and Idd8 (Chr 14) did not segregate with IDDM in either the BC1 or the F2 generation. Diabetogenic NOD-derived alleles at Idd2 (Chr 9), Idd3 (Chr 3), and Idd10 (Chr 3) were segregating in the BC1. An NON-derived allele contributing to susceptibility on Chr 7 (Idd7) was also detected. Dominant traits, detectable only in the F2 cross, were encoded by Chr 4 (Idd9) and two newly mapped loci on Chr 13 (Idd14) and 5 (Idd15). A third dominant trait was encoded by Chr 6 (possibly Idd6), but here, in contrast to Idd9, Idd14, and Idd15, the NON allele was diabetogenic. Stepwise logistic regression analysis of the BC1 and F2 data confirmed that the ability to identify certainty of the non-MHC Idd loci was contingent on the extent of homozygosity for NOD background genes. This study shows that the diabetogenic phenotype can be achieved through the actions of variable combinations of MHC-unlinked genes and a diabetogenic MHC haplotype.


Journal of Leukocyte Biology | 2003

Antibody-mediated blockade of the CXCR3 chemokine receptor results in diminished recruitment of T helper 1 cells into sites of inflammation

Jenny H. Xie; Naomi Nomura; Min Lu; Shiow-Ling Chen; Greg Koch; Youmin Weng; Ray Rosa; Jerry Di Salvo; John S. Mudgett; Laurence B. Peterson; Linda S. Wicker; Julie A. DeMartino

Naïve T cells, when activated by specific antigen and cytokines, up‐regulate adhesion molecules as well as chemokine receptors on their surface, which allows them to migrate to inflamed tissues. Human studies have shown that CXCR3 is one of the chemokine receptors that is induced during T cell activation. Moreover, CXCR3‐positive T cells are enriched at inflammatory sites in patients with autoimmune diseases such as rheumatoid arthritis and multiple sclerosis. In this study, we use a mouse model of inflammation to demonstrate that CXCR3 is required for activated T cell transmigration to inflamed tissue. Using an anti‐ mCXCR3 antibody, we have shown that in vitro‐differentiated T helper (Th) 1 and Th2 cells up‐regulated CXCR3 upon stimulation with specific antigen/major histocompatibility complex. However, only Th1 cells, when adoptively transferred to syngeneic recipients, are efficiently recruited to the peritoneum in an adjuvant‐induced peritonitis model. Furthermore, the neutralizing anti‐mCXCR3 antibody profoundly inhibits the recruitment of Th1 cells to the inflamed peritoneum. Real‐time, quantitative reverse transcriptase‐polymerase chain reaction analysis demonstrates that the CXCR3 ligands, interferon (IFN)‐inducible protein 10 (CXCL10) and IFN‐inducible T cell α chemoattractant (CXCL11), are among the many chemokines induced in the adjuvant‐treated peritoneum. The anti‐mCXCR3 antibody is also effective in inhibiting a delayed‐type hypersensitivity response, which is largely mediated by enhanced trafficking of activated T cells to peripheral inflammatory sites. Collectively, our results suggest that CXCR3 has a critical role in T cell transmigration to sites of inflammation and thus, may serve as a molecular target for anti‐inflammatory therapies.


Diabetes | 1997

Mapping of the IDDM locus Idd3 to a 0.35-cM interval containing the interleukin-2 gene.

Paul W. Denny; Christopher J. Lord; Natasha Hill; Juliet V. Goy; Elaine R. Levy; Patricia L Podolin; Laurence B. Peterson; Linda S. Wicker; John A. Todd; Paul A. Lyons

Currently, 16 loci that contribute to the development of IDDM in the NOD mouse have been mapped by linkage analysis. To fine map these loci, we used congenic mapping. Using this approach, we localized the Idd3 locus to a 0.35-cM interval on chromosome 3 containing the Il2 gene. Segregation analysis of the known variations within this interval indicated that only one variant, a serine-to-proline substitution at position 6 of the mature interleukin-2 (IL-2) protein, consistently segregates with IDDM in crosses between NOD and a series of nondiabetic mouse strains. These data, taken together with the immunomodulatory role of IL-2, provide circumstantial evidence in support of the hypothesis that Idd3 is an allelic variation of the Il2 gene, or a variant in strong linkage disequilibrium.

Collaboration


Dive into the Laurence B. Peterson's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

John A. Todd

Wellcome Trust Centre for Human Genetics

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kara Hunter

University of Cambridge

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christopher J. Lord

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David V. Serreze

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge