Network


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

Hotspot


Dive into the research topics where Lisa R. Forbes is active.

Publication


Featured researches published by Lisa R. Forbes.


JAMA | 2014

Newborn Screening for Severe Combined Immunodeficiency in 11 Screening Programs in the United States

Antonia Kwan; Roshini S. Abraham; Robert Currier; Amy Brower; Karen Andruszewski; Jordan K. Abbott; Mei W. Baker; Mark Ballow; Louis Bartoshesky; Francisco A. Bonilla; Charles D. Brokopp; Edward G. Brooks; Michele Caggana; Jocelyn Celestin; Joseph A. Church; Anne Marie Comeau; James A. Connelly; Morton J. Cowan; Charlotte Cunningham-Rundles; Trivikram Dasu; Nina Dave; Maria Teresa De La Morena; Ulrich A. Duffner; Chin To Fong; Lisa R. Forbes; Debra Freedenberg; Erwin W. Gelfand; Jaime E. Hale; I. Celine Hanson; Beverly N. Hay

IMPORTANCE Newborn screening for severe combined immunodeficiency (SCID) using assays to detect T-cell receptor excision circles (TRECs) began in Wisconsin in 2008, and SCID was added to the national recommended uniform panel for newborn screened disorders in 2010. Currently 23 states, the District of Columbia, and the Navajo Nation conduct population-wide newborn screening for SCID. The incidence of SCID is estimated at 1 in 100,000 births. OBJECTIVES To present data from a spectrum of SCID newborn screening programs, establish population-based incidence for SCID and other conditions with T-cell lymphopenia, and document early institution of effective treatments. DESIGN Epidemiological and retrospective observational study. SETTING Representatives in states conducting SCID newborn screening were invited to submit their SCID screening algorithms, test performance data, and deidentified clinical and laboratory information regarding infants screened and cases with nonnormal results. Infants born from the start of each participating program from January 2008 through the most recent evaluable date prior to July 2013 were included. Representatives from 10 states plus the Navajo Area Indian Health Service contributed data from 3,030,083 newborns screened with a TREC test. MAIN OUTCOMES AND MEASURES Infants with SCID and other diagnoses of T-cell lymphopenia were classified. Incidence and, where possible, etiologies were determined. Interventions and survival were tracked. RESULTS Screening detected 52 cases of typical SCID, leaky SCID, and Omenn syndrome, affecting 1 in 58,000 infants (95% CI, 1/46,000-1/80,000). Survival of SCID-affected infants through their diagnosis and immune reconstitution was 87% (45/52), 92% (45/49) for infants who received transplantation, enzyme replacement, and/or gene therapy. Additional interventions for SCID and non-SCID T-cell lymphopenia included immunoglobulin infusions, preventive antibiotics, and avoidance of live vaccines. Variations in definitions and follow-up practices influenced the rates of detection of non-SCID T-cell lymphopenia. CONCLUSIONS AND RELEVANCE Newborn screening in 11 programs in the United States identified SCID in 1 in 58,000 infants, with high survival. The usefulness of detection of non-SCID T-cell lymphopenias by the same screening remains to be determined.


Blood | 2015

Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations.

Joshua D. Milner; Tiphanie P. Vogel; Lisa R. Forbes; Chi A. Ma; Asbjørg Stray-Pedersen; Julie E. Niemela; Jonathan J. Lyons; Karin R. Engelhardt; Yu Zhang; Nermina Topcagic; Elisha D. O. Roberson; Helen F. Matthews; James W. Verbsky; Trivikram Dasu; Alexander Vargas-Hernández; Nidhy P. Varghese; Kenneth L. McClain; Lina Karam; Karen Nahmod; George Makedonas; Emily M. Mace; Hanne Sørmo Sorte; Gøri Perminow; V. Koneti Rao; Michael P. O’Connell; Susan Price; Helen C. Su; Morgan Butrick; Joshua McElwee; Jason D. Hughes

Germline loss-of-function mutations in the transcription factor signal transducer and activator of transcription 3 (STAT3) cause immunodeficiency, whereas somatic gain-of-function mutations in STAT3 are associated with large granular lymphocytic leukemic, myelodysplastic syndrome, and aplastic anemia. Recently, germline mutations in STAT3 have also been associated with autoimmune disease. Here, we report on 13 individuals from 10 families with lymphoproliferation and early-onset solid-organ autoimmunity associated with 9 different germline heterozygous mutations in STAT3. Patients exhibited a variety of clinical features, with most having lymphadenopathy, autoimmune cytopenias, multiorgan autoimmunity (lung, gastrointestinal, hepatic, and/or endocrine dysfunction), infections, and short stature. Functional analyses demonstrate that these mutations confer a gain-of-function in STAT3 leading to secondary defects in STAT5 and STAT1 phosphorylation and the regulatory T-cell compartment. Treatment targeting a cytokine pathway that signals through STAT3 led to clinical improvement in 1 patient, suggesting a potential therapeutic option for such patients. These results suggest that there is a broad range of autoimmunity caused by germline STAT3 gain-of-function mutations, and that hematologic autoimmunity is a major component of this newly described disorder. Some patients for this study were enrolled in a trial registered at www.clinicaltrials.gov as #NCT00001350.


Journal of Clinical Investigation | 2012

Human immunodeficiency-causing mutation defines CD16 in spontaneous NK cell cytotoxicity

Jennifer T. Grier; Lisa R. Forbes; Linda Monaco-Shawver; Jennifer Oshinsky; T. Prescott Atkinson; Curtis T. Moody; Rahul Pandey; Kerry S. Campbell; Jordan S. Orange

The Fc receptor on NK cells, FcγRIIIA (CD16), has been extensively studied for its role in mediating antibody-dependent cellular cytotoxicity (ADCC). A homozygous missense mutation in CD16 (encoding a L66H substitution) is associated with severe herpesvirus infections in rare patients. Here, we identified a new patient with this CD16 mutation and compared the patients NK cells to those of the originally reported patient. Patients with the L66H mutation had intact ADCC, but deficient spontaneous NK cell cytotoxicity and decreased surface expression of CD2, a coactivation receptor. Mechanistic studies in a human NK cell line, NK-92, demonstrated that CD16 expression correlated with CD2 surface levels and enabled killing of a melanoma cell line typically resistant to CD16-deficient NK-92 cells. An association between CD16 and CD2 was identified biochemically and at the immunological synapse, which elicited CD16 signaling after CD2 engagement. Stable expression of CD16 L66H in NK-92 cells recapitulated the patient phenotype, abrogating association of CD16 with CD2 as well as CD16 signaling after CD2 ligation. Thus, CD16 serves a role in NK cell-mediated spontaneous cytotoxicity through a specific association with CD2 and represents a potential mechanism underlying a human congenital immunodeficiency.


Immunology and Cell Biology | 2014

Cell biological steps and checkpoints in accessing NK cell cytotoxicity

Emily M. Mace; Prachi Dongre; Hsiang-Ting Hsu; Papiya Sinha; Ashley Mentlik James; Shaina S. Mann; Lisa R. Forbes; Levi B. Watkin; Jordan S. Orange

Natural killer (NK) cell‐mediated cytotoxicity is governed by the formation of a lytic immune synapse in discrete regulated steps, which give rise to an extensive array of cellular checkpoints in accessing NK cell‐mediated cytolytic defense. Appropriate progression through these cell biological steps is critical for the directed secretion of specialized secretory lysosomes and subsequent target cell death. Here we highlight recent discoveries in the formation of the NK cell cytolytic synapse as well as the molecular steps and cell biological checkpoints required for this essential host defense process.


Nature Genetics | 2015

COPA mutations impair ER-Golgi transport and cause hereditary autoimmune-mediated lung disease and arthritis

Levi B. Watkin; Birthe Jessen; Wojciech Wiszniewski; Timothy J. Vece; Max Jan; Youbao Sha; Maike Thamsen; Regie Lyn P. Santos-Cortez; Kwanghyuk Lee; Tomasz Gambin; Lisa R. Forbes; Christopher S. Law; Asbjørg Stray-Pedersen; Mickie H. Cheng; Emily M. Mace; Mark S. Anderson; Dongfang Liu; Ling Fung Tang; Sarah K. Nicholas; Karen Nahmod; George Makedonas; Debra L. Canter; Pui-Yan Kwok; John Hicks; Kirk D. Jones; Samantha Penney; Shalini N. Jhangiani; Michael D. Rosenblum; Sharon D. Dell; Michael Waterfield

Unbiased genetic studies have uncovered surprising molecular mechanisms in human cellular immunity and autoimmunity. We performed whole-exome sequencing and targeted sequencing in five families with an apparent mendelian syndrome of autoimmunity characterized by high-titer autoantibodies, inflammatory arthritis and interstitial lung disease. We identified four unique deleterious variants in the COPA gene (encoding coatomer subunit α) affecting the same functional domain. Hypothesizing that mutant COPA leads to defective intracellular transport via coat protein complex I (COPI), we show that COPA variants impair binding to proteins targeted for retrograde Golgi-to-ER transport. Additionally, expression of mutant COPA results in ER stress and the upregulation of cytokines priming for a T helper type 17 (TH17) response. Patient-derived CD4+ T cells also demonstrate significant skewing toward a TH17 phenotype that is implicated in autoimmunity. Our findings uncover an unexpected molecular link between a vesicular transport protein and a syndrome of autoimmunity manifested by lung and joint disease.


The Journal of Allergy and Clinical Immunology | 2017

Primary immunodeficiency diseases: Genomic approaches delineate heterogeneous Mendelian disorders

Asbjørg Stray-Pedersen; Hanne Sørmo Sorte; Pubudu Saneth Samarakoon; Tomasz Gambin; Ivan K. Chinn; Zeynep Coban Akdemir; Hans Christian Erichsen; Lisa R. Forbes; Shen Gu; Bo Yuan; Shalini N. Jhangiani; Donna M. Muzny; Olaug K. Rødningen; Ying Sheng; Sarah K. Nicholas; Lenora M. Noroski; Filiz O. Seeborg; Carla M. Davis; Debra L. Canter; Emily M. Mace; Timothy J. Vece; Carl E. Allen; Harshal Abhyankar; Philip M. Boone; Christine R. Beck; Wojciech Wiszniewski; Børre Fevang; Pål Aukrust; Geir E. Tjønnfjord; Tobias Gedde-Dahl

Background: Primary immunodeficiency diseases (PIDDs) are clinically and genetically heterogeneous disorders thus far associated with mutations in more than 300 genes. The clinical phenotypes derived from distinct genotypes can overlap. Genetic etiology can be a prognostic indicator of disease severity and can influence treatment decisions. Objective: We sought to investigate the ability of whole‐exome screening methods to detect disease‐causing variants in patients with PIDDs. Methods: Patients with PIDDs from 278 families from 22 countries were investigated by using whole‐exome sequencing. Computational copy number variant (CNV) prediction pipelines and an exome‐tiling chromosomal microarray were also applied to identify intragenic CNVs. Analytic approaches initially focused on 475 known or candidate PIDD genes but were nonexclusive and further tailored based on clinical data, family history, and immunophenotyping. Results: A likely molecular diagnosis was achieved in 110 (40%) unrelated probands. Clinical diagnosis was revised in about half (60/110) and management was directly altered in nearly a quarter (26/110) of families based on molecular findings. Twelve PIDD‐causing CNVs were detected, including 7 smaller than 30 Kb that would not have been detected with conventional diagnostic CNV arrays. Conclusion: This high‐throughput genomic approach enabled detection of disease‐related variants in unexpected genes; permitted detection of low‐grade constitutional, somatic, and revertant mosaicism; and provided evidence of a mutational burden in mixed PIDD immunophenotypes.


Clinical and Experimental Immunology | 2014

Vaccine-associated varicella and rubella infections in severe combined immunodeficiency with isolated CD4 lymphocytopenia and mutations in IL7R detected by tandem whole exome sequencing and chromosomal microarray

D. K. Bayer; Caridad Martinez; Hanne Sørmo Sorte; Lisa R. Forbes; G. J. Demmler-Harrison; I. C. Hanson; N. M. Pearson; Lenora M. Noroski; S. R. Zaki; W. J. Bellini; Magalie S. Leduc; Yaping Yang; Christine M. Eng; Ankita Patel; Olaug K. Rødningen; Donna M. Muzny; Richard A. Gibbs; Ian M. Campbell; Chad A. Shaw; M. W. Baker; Victor Wei Zhang; James R. Lupski; Jordan S. Orange; Filiz O. Seeborg; Asbjørg Stray-Pedersen

In areas without newborn screening for severe combined immunodeficiency (SCID), disease‐defining infections may lead to diagnosis, and in some cases, may not be identified prior to the first year of life. We describe a female infant who presented with disseminated vaccine‐acquired varicella (VZV) and vaccine‐acquired rubella infections at 13 months of age. Immunological evaluations demonstrated neutropenia, isolated CD4 lymphocytopenia, the presence of CD8+ T cells, poor lymphocyte proliferation, hypergammaglobulinaemia and poor specific antibody production to VZV infection and routine immunizations. A combination of whole exome sequencing and custom‐designed chromosomal microarray with exon coverage of primary immunodeficiency genes detected compound heterozygous mutations (one single nucleotide variant and one intragenic copy number variant involving one exon) within the IL7R gene. Mosaicism for wild‐type allele (20–30%) was detected in pretransplant blood and buccal DNA and maternal engraftment (5–10%) demonstrated in pretransplant blood DNA. This may be responsible for the patients unusual immunological phenotype compared to classical interleukin (IL)‐7Rα deficiency. Disseminated VZV was controlled with anti‐viral and immune‐based therapy, and umbilical cord blood stem cell transplantation was successful. Retrospectively performed T cell receptor excision circle (TREC) analyses completed on neonatal Guthrie cards identified absent TREC. This case emphasizes the danger of live viral vaccination in severe combined immunodeficiency (SCID) patients and the importance of newborn screening to identify patients prior to high‐risk exposures. It also illustrates the value of aggressive pathogen identification and treatment, the influence newborn screening can have on morbidity and mortality and the significant impact of newer genomic diagnostic tools in identifying the underlying genetic aetiology for SCID patients.


The Journal of Allergy and Clinical Immunology | 2017

Ruxolitinib reverses dysregulated T helper cell responses and controls autoimmunity caused by a novel signal transducer and activator of transcription 1 (STAT1) gain-of-function mutation

Katja G. Weinacht; Louis-Marie Charbonnier; Fayhan Alroqi; Ashley S. Plant; Qi Qiao; Hao Wu; Clement Ma; Troy R. Torgerson; Sergio D. Rosenzweig; Thomas A. Fleisher; Luigi D. Notarangelo; Imelda C. Hanson; Lisa R. Forbes; Talal A. Chatila

Background: Gain‐of‐function (GOF) mutations in the human signal transducer and activator of transcription 1 (STAT1) manifest in immunodeficiency and autoimmunity with impaired TH17 cell differentiation and exaggerated responsiveness to type I and II interferons. Allogeneic bone marrow transplantation has been attempted in severely affected patients, but outcomes have been poor. Objective: We sought to define the effect of increased STAT1 activity on T helper cell polarization and to investigate the therapeutic potential of ruxolitinib in treating autoimmunity secondary to STAT1 GOF mutations. Methods: We used in vitro polarization assays, as well as phenotypic and functional analysis of STAT1‐mutated patient cells. Results: We report a child with a novel mutation in the linker domain of STAT1 who had life‐threatening autoimmune cytopenias and chronic mucocutaneous candidiasis. Naive lymphocytes from the affected patient displayed increased TH1 and follicular T helper cell and suppressed TH17 cell responses. The mutation augmented cytokine‐induced STAT1 phosphorylation without affecting dephosphorylation kinetics. Treatment with the Janus kinase 1/2 inhibitor ruxolitinib reduced hyperresponsiveness to type I and II interferons, normalized TH1 and follicular T helper cell responses, improved TH17 differentiation, cured mucocutaneous candidiasis, and maintained remission of immune‐mediated cytopenias. Conclusions: Autoimmunity and infection caused by STAT1 GOF mutations are the result of dysregulated T helper cell responses. Janus kinase inhibitor therapy could represent an effective targeted treatment for long‐term disease control in severely affected patients for whom hematopoietic stem cell transplantation is not available.


Immunologic Research | 2015

Practical NK cell phenotyping and variability in healthy adults

Laura S. Angelo; Pinaki P. Banerjee; Linda Monaco-Shawver; Joshua B. Rosen; George Makedonas; Lisa R. Forbes; Emily M. Mace; Jordan S. Orange

Human natural killer (NK) cells display a wide array of surface and intracellular markers that indicate various states of differentiation and/or levels of effector function. These NK cell subsets exist simultaneously in peripheral blood and may vary among individuals. We examined variety among selected NK cell receptors expressed by NK cells from normal donors, as well as the distribution of select NK cell subsets and NK cell receptor expression over time in several individual donors. Peripheral blood mononuclear cells were evaluated using flow cytometry via fluorochrome-conjugated antibodies against a number of NK cell receptors. Results were analyzed for both mean fluorescence intensity (MFI) and the percent positive cells for each receptor. CD56bright and CD56dim NK cell subsets were also considered separately, as was variation in receptor expression in NK cell subsets over time in selected individuals. Through this effort, we provide ranges of NK cell surface receptor expression for a local adult population as well as provide insight into intra-individual variation.


Journal of Clinical Immunology | 2016

Copa Syndrome: a Novel Autosomal Dominant Immune Dysregulatory Disease

Timothy J. Vece; Levi B. Watkin; Sarah K. Nicholas; Debra Canter; Michael C. Braun; Robert Paul Guillerman; Karen W. Eldin; Grant Bertolet; Scott D. McKinley; Marietta M. de Guzman; Lisa R. Forbes; Ivan K. Chinn; Jordan S. Orange

Inherently defective immunity typically results in either ineffective host defense, immune regulation, or both. As a category of primary immunodeficiency diseases, those that impair immune regulation can lead to autoimmunity and/or autoinflammation. In this review we focus on one of the most recently discovered primary immunodeficiencies that leads to immune dysregulation: “Copa syndrome”. Copa syndrome is named for the gene mutated in the disease, which encodes the alpha subunit of the coatomer complex-I that, in aggregate, is devoted to transiting molecular cargo from the Golgi complex to the endoplasmic reticulum (ER). Copa syndrome is autosomal dominant with variable expressivity and results from mutations affecting a narrow amino acid stretch in the COPA gene-encoding COPα protein. Patients with these mutations typically develop arthritis and interstitial lung disease with pulmonary hemorrhage representing a striking feature. Immunologically Copa syndrome is associated with autoantibody development, increased Th17 cells and pro-inflammatory cytokine expression including IL-1β and IL-6. Insights have also been gained into the underlying mechanism of Copa syndrome, which include excessive ER stress owing to the impaired return of proteins from the Golgi, and presumably resulting aberrant cellular autophagy. As such it represents a novel cellular disorder of intracellular trafficking associated with a specific clinical presentation and phenotype.

Collaboration


Dive into the Lisa R. Forbes's collaboration.

Top Co-Authors

Avatar

Jordan S. Orange

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Emily M. Mace

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Caridad Martinez

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Ivan K. Chinn

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Imelda C. Hanson

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Sarah K. Nicholas

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Robert A. Krance

Center for Cell and Gene Therapy

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Filiz O. Seeborg

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Helen E. Heslop

Center for Cell and Gene Therapy

View shared research outputs
Researchain Logo
Decentralizing Knowledge