Network


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

Hotspot


Dive into the research topics where Kerry Dobbs is active.

Publication


Featured researches published by Kerry Dobbs.


Journal of Experimental Medicine | 2015

Human HOIP and LUBAC deficiency underlies autoinflammation, immunodeficiency, amylopectinosis, and lymphangiectasia

Bertrand Boisson; Emmanuel Laplantine; Kerry Dobbs; Aurélie Cobat; Nadine Tarantino; Melissa Hazen; Hart G.W. Lidov; Gregory Hopkins; Likun Du; Aziz Belkadi; Maya Chrabieh; Yuval Itan; Capucine Picard; Jean-Christophe Fournet; Hermann Eibel; Erdyni Tsitsikov; Sung-Yun Pai; Laurent Abel; Waleed Al-Herz; Jean-Laurent Casanova; Alain Israël; Luigi D. Notarangelo

Boisson et al. report a human homozygous mutation of HOIP, the gene encoding the catalytic component of the linear ubiquitination chain assembly complex, LUBAC. The missense alleles impair the expression of HOIP, destabilizing the LUBAC complex and resulting in immune cell dysfunction leading to multiorgan inflammation, combined immunodeficiency, subclinical amylopectinosis, and systemic lymphangiectactasia.


The Journal of Allergy and Clinical Immunology | 2013

Whole-exome sequencing identifies tetratricopeptide repeat domain 7A (TTC7A) mutations for combined immunodeficiency with intestinal atresias

Rui Chen; Silvia Giliani; Gaetana Lanzi; George Mias; Silvia Lonardi; Kerry Dobbs; John P. Manis; Hogune Im; Jennifer E.G. Gallagher; Douglas H. Phanstiel; Ghia Euskirchen; Philippe Lacroute; Keith Bettinger; Daniele Moratto; Katja G. Weinacht; Davide Montin; Eleonora Gallo; Giovanna Mangili; Fulvio Porta; Lucia Dora Notarangelo; Stefania Pedretti; Waleed Al-Herz; Anne Marie Comeau; Russell S. Traister; Sung-Yun Pai; Graziella Carella; Fabio Facchetti; Kari C. Nadeau; Michael Snyder; Luigi D. Notarangelo

BACKGROUND Combined immunodeficiency with multiple intestinal atresias (CID-MIA) is a rare hereditary disease characterized by intestinal obstructions and profound immune defects. OBJECTIVE We sought to determine the underlying genetic causes of CID-MIA by analyzing the exomic sequences of 5 patients and their healthy direct relatives from 5 unrelated families. METHODS We performed whole-exome sequencing on 5 patients with CID-MIA and 10 healthy direct family members belonging to 5 unrelated families with CID-MIA. We also performed targeted Sanger sequencing for the candidate gene tetratricopeptide repeat domain 7A (TTC7A) on 3 additional patients with CID-MIA. RESULTS Through analysis and comparison of the exomic sequence of the subjects from these 5 families, we identified biallelic damaging mutations in the TTC7A gene, for a total of 7 distinct mutations. Targeted TTC7A gene sequencing in 3 additional unrelated patients with CID-MIA revealed biallelic deleterious mutations in 2 of them, as well as an aberrant splice product in the third patient. Staining of normal thymus showed that the TTC7A protein is expressed in thymic epithelial cells, as well as in thymocytes. Moreover, severe lymphoid depletion was observed in the thymus and peripheral lymphoid tissues from 2 patients with CID-MIA. CONCLUSIONS We identified deleterious mutations of the TTC7A gene in 8 unrelated patients with CID-MIA and demonstrated that the TTC7A protein is expressed in the thymus. Our results strongly suggest that TTC7A gene defects cause CID-MIA.


Journal of Clinical Investigation | 2015

Broad-spectrum antibodies against self-antigens and cytokines in RAG deficiency

Jolan E. Walter; Lindsey B. Rosen; Krisztian Csomos; Jacob Rosenberg; Divij Mathew; Marton Keszei; Boglarka Ujhazi; Karin Chen; Yu Nee Lee; Irit Tirosh; Kerry Dobbs; Waleed Al-Herz; Morton J. Cowan; Jennifer M. Puck; Jack Bleesing; Michael Grimley; Harry L. Malech; Suk See De Ravin; Andrew R. Gennery; Roshini S. Abraham; Avni Y. Joshi; Thomas G. Boyce; Manish J. Butte; Kari C. Nadeau; Imelda Balboni; Kathleen E. Sullivan; Javeed Akhter; Mehdi Adeli; Reem Elfeky; Dalia H. El-Ghoneimy

Patients with mutations of the recombination-activating genes (RAG) present with diverse clinical phenotypes, including severe combined immune deficiency (SCID), autoimmunity, and inflammation. However, the incidence and extent of immune dysregulation in RAG-dependent immunodeficiency have not been studied in detail. Here, we have demonstrated that patients with hypomorphic RAG mutations, especially those with delayed-onset combined immune deficiency and granulomatous/autoimmune manifestations (CID-G/AI), produce a broad spectrum of autoantibodies. Neutralizing anti-IFN-α or anti-IFN-ω antibodies were present at detectable levels in patients with CID-G/AI who had a history of severe viral infections. As this autoantibody profile is not observed in a wide range of other primary immunodeficiencies, we hypothesized that recurrent or chronic viral infections may precipitate or aggravate immune dysregulation in RAG-deficient hosts. We repeatedly challenged Rag1S723C/S723C mice, which serve as a model of leaky SCID, with agonists of the virus-recognizing receptors TLR3/MDA5, TLR7/-8, and TLR9 and found that this treatment elicits autoantibody production. Altogether, our data demonstrate that immune dysregulation is an integral aspect of RAG-associated immunodeficiency and indicate that environmental triggers may modulate the phenotypic expression of autoimmune manifestations.


The New England Journal of Medicine | 2015

Inherited DOCK2 Deficiency in Patients with Early-Onset Invasive Infections

Abstr Act; Kerry Dobbs; C. Domínguez Conde; Silvia Parolini; M. Audry; Janet Chou; E. Haapaniemi; Sevgi Keles; Ivan Bilic; Satoshi Okada; S. Rounioja; Kelly Capuder; Kerstin Felgentreff; V. Pedergnana; Bertrand Boisson; A. Ensari; Michael Schuster; Alessandro Moretta; Yuval Itan; Ornella Patrizi; Flore Rozenberg; Pierre Lebon; J. Saarela; Mikael Knip; Slavé Petrovski; B. Savas; Axel Schambach; Giovanna Tabellini; Christoph Bock; Laurent Abel

Background Combined immunodeficiencies are marked by inborn errors of T-cell immunity in which the T cells that are present are quantitatively or functionally deficient. Impaired humoral immunity is also common. Patients have severe infections, autoimmunity, or both. The specific molecular, cellular, and clinical features of many types of combined immunodeficiencies remain unknown. Methods We performed genetic and cellular immunologic studies involving five unrelated children with early-onset invasive bacterial and viral infections, lymphopenia, and defective T-cell, B-cell, and natural killer (NK)-cell responses. Two patients died early in childhood; after allogeneic hematopoietic stem-cell transplantation, the other three had normalization of T-cell function and clinical improvement. Results We identified biallelic mutations in the dedicator of cytokinesis 2 gene (DOCK2) in these five patients. RAC1 activation was impaired in the T cells. Chemokine-induced migration and actin polymerization were defective in the T cells, B cells, and NK cells. NK-cell degranulation was also affected. Interferon-α and interferon-λ production by peripheral-blood mononuclear cells was diminished after viral infection. Moreover, in DOCK2-deficient fibroblasts, viral replication was increased and virus-induced cell death was enhanced; these conditions were normalized by treatment with interferon alfa-2b or after expression of wild-type DOCK2. Conclusions Autosomal recessive DOCK2 deficiency is a new mendelian disorder with pleiotropic defects of hematopoietic and nonhematopoietic immunity. Children with clinical features of combined immunodeficiencies, especially with early-onset, invasive infections, may have this condition. (Supported by the National Institutes of Health and others.).


Proceedings of the National Academy of Sciences of the United States of America | 2014

Differential role of nonhomologous end joining factors in the generation, DNA damage response, and myeloid differentiation of human induced pluripotent stem cells

Kerstin Felgentreff; Likun Du; Katja G. Weinacht; Kerry Dobbs; Margarita Bartish; Silvia Giliani; Thorsten M. Schlaeger; Alexander L. DeVine; Axel Schambach; Lisa Woodbine; Graham Davies; Sachin N. Baxi; Mirjam van der Burg; Jack Bleesing; Andrew R. Gennery; John P. Manis; Qiang Pan-Hammarström; Luigi D. Notarangelo

Significance In this study, we compare the differential impact of nonhomologous end-joining–deficient induced pluripotent stem cells (iPSCs) derived from patients with mutations in genes encoding DNA ligase 4, DNA-protein kinase catalytic subunit, or Artemis, on cellular reprogramming, DNA damage repair, and myeloid differentiation. We found a predominant role for ligase 4 in genomic stability and myeloid differentiation of iPSCs, but a minor one for Artemis. Disease modeling with patient-derived iPSCs may provide significant insights into phenotypic variability associated with mutations in various genes along the same pathway and may serve as a platform to explore tissue-specific consequences of single gene defects and development of therapeutic interventions. Nonhomologous end-joining (NHEJ) is a key pathway for efficient repair of DNA double-strand breaks (DSBs) and V(D)J recombination. NHEJ defects in humans cause immunodeficiency and increased cellular sensitivity to ionizing irradiation (IR) and are variably associated with growth retardation, microcephaly, and neurodevelopmental delay. Repair of DNA DSBs is important for reprogramming of somatic cells into induced pluripotent stem cells (iPSCs). To compare the specific contribution of DNA ligase 4 (LIG4), Artemis, and DNA-protein kinase catalytic subunit (PKcs) in this process and to gain insights into phenotypic variability associated with these disorders, we reprogrammed patient-derived fibroblast cell lines with NHEJ defects. Deficiencies of LIG4 and of DNA-PK catalytic activity, but not Artemis deficiency, were associated with markedly reduced reprogramming efficiency, which could be partially rescued by genetic complementation. Moreover, we identified increased genomic instability in LIG4-deficient iPSCs. Cell cycle synchronization revealed a severe defect of DNA repair and a G0/G1 cell cycle arrest, particularly in LIG4- and DNA-PK catalytically deficient iPSCs. Impaired myeloid differentiation was observed in LIG4-, but not Artemis- or DNA-PK–mutated iPSCs. These results indicate a critical importance of the NHEJ pathway for somatic cell reprogramming, with a major role for LIG4 and DNA-PKcs and a minor, if any, for Artemis.


Frontiers in Immunology | 2014

Next Generation Sequencing Reveals Skewing of the T and B Cell Receptor Repertoires in Patients with Wiskott–Aldrich Syndrome

Amy E. O’Connell; Stefano Volpi; Kerry Dobbs; Claudia Fiorini; Erdyni Tsitsikov; Helen de Boer; Isil B. Barlan; Jenny M. Despotovic; Francisco J. Espinosa-Rosales; I. Celine Hanson; Maria Kanariou; Roxana Martínez-Beckerat; Alvaro Mayorga-Sirera; Carmen Mejia-Carvajal; Nesrine Radwan; Aaron R. Weiss; Sung-Yun Pai; Yu Nee Lee; Luigi D. Notarangelo

The Wiskott–Aldrich syndrome (WAS) is due to mutations of the WAS gene encoding for the cytoskeletal WAS protein, leading to abnormal downstream signaling from the T cell and B cell antigen receptors (TCR and BCR). We hypothesized that the impaired signaling through the TCR and BCR in WAS would subsequently lead to aberrations in the immune repertoire of WAS patients. Using next generation sequencing (NGS), the T cell receptor β and B cell immunoglobulin heavy chain (IGH) repertoires of eight patients with WAS and six controls were sequenced. Clonal expansions were identified within memory CD4+ cells as well as in total, naïve and memory CD8+ cells from WAS patients. In the B cell compartment, WAS patient IGH repertoires were also clonally expanded and showed skewed usage of IGHV and IGHJ genes, and increased usage of IGHG constant genes, compared with controls. To our knowledge, this is the first study that demonstrates significant abnormalities of the immune repertoire in WAS patients using NGS.


Journal of Experimental Medicine | 2017

EXTL3 mutations cause skeletal dysplasia, immune deficiency, and developmental delay

Stefano Volpi; Yasuhiro Yamazaki; Patrick M. Brauer; Ellen van Rooijen; Atsuko Hayashida; Anne Slavotinek; Hye Sun Kuehn; Maja Di Rocco; Carlo Rivolta; Ileana Bortolomai; Likun Du; Kerstin Felgentreff; Lisa Ott de Bruin; Kazutaka Hayashida; George Freedman; Genni Enza Marcovecchio; Kelly Capuder; Prisni Rath; Nicole Luche; Elliott J. Hagedorn; Antonella Buoncompagni; Beryl Royer-Bertrand; Silvia Giliani; Pietro Luigi Poliani; Luisa Imberti; Kerry Dobbs; Fabienne E. Poulain; Alberto Martini; John P. Manis; Robert J. Linhardt

We studied three patients with severe skeletal dysplasia, T cell immunodeficiency, and developmental delay. Whole-exome sequencing revealed homozygous missense mutations affecting exostosin-like 3 (EXTL3), a glycosyltransferase involved in heparan sulfate (HS) biosynthesis. Patient-derived fibroblasts showed abnormal HS composition and altered fibroblast growth factor 2 signaling, which was rescued by overexpression of wild-type EXTL3 cDNA. Interleukin-2–mediated STAT5 phosphorylation in patients’ lymphocytes was markedly reduced. Interbreeding of the extl3-mutant zebrafish (box) with Tg(rag2:green fluorescent protein) transgenic zebrafish revealed defective thymopoiesis, which was rescued by injection of wild-type human EXTL3 RNA. Targeted differentiation of patient-derived induced pluripotent stem cells showed a reduced expansion of lymphohematopoietic progenitor cells and defects of thymic epithelial progenitor cell differentiation. These data identify EXTL3 mutations as a novel cause of severe immune deficiency with skeletal dysplasia and developmental delay and underline a crucial role of HS in thymopoiesis and skeletal and brain development.


Science immunology | 2016

Characterization of T and B cell repertoire diversity in patients with RAG deficiency

Yu Nee Lee; Francesco Frugoni; Kerry Dobbs; Irit Tirosh; Likun Du; Francesca Ververs; Heng Ru; L. de Bruin; M. Adeli; Jacob Bleesing; David Buchbinder; Manish J. Butte; Caterina Cancrini; Karin Chen; S. Choo; Reem Elfeky; Andrea Finocchi; Ramsay L. Fuleihan; Andrew R. Gennery; Dalia H. El-Ghoneimy; Lauren A. Henderson; Waleed Al-Herz; Elham Hossny; Robert P. Nelson; Sung-Yun Pai; Niraj C. Patel; Shereen M. Reda; P. Soler-Palacin; Raz Somech; Paolo Palma

Differences in B and T cell repertoires in patients with RAG deficiency associate with clinical severity. Taking SCID genetics to the clinic Mutations that lead to deficiencies in the recombination-activating genes RAG1 and RAG2 result in a spectrum of immunodeficiencies ranging from loss of T and/or B cell repertoire diversity to a complete lack of T and B cells—severe combined immunodeficiency (SCID). Here, Lee et al. perform next-generation B and T cell repertoire sequencing on 12 patients with RAG mutations who have immunodeficiencies of varying severity. They found that the level of repertoire skewing was associated with the severity of disease and that specific repertoire deficiencies were associated with particular phenotypes. These data support a genotype-phenotype connection for primary immunodeficiencies. Recombination-activating genes 1 and 2 (RAG1 and RAG2) play a critical role in T and B cell development by initiating the recombination process that controls the expression of T cell receptor (TCR) and immunoglobulin genes. Mutations in the RAG1 and RAG2 genes in humans cause a broad spectrum of phenotypes, including severe combined immunodeficiency (SCID) with lack of T and B cells, Omenn syndrome, leaky SCID, and combined immunodeficiency with granulomas or autoimmunity (CID-G/AI). Using next-generation sequencing, we analyzed the TCR and B cell receptor (BCR) repertoire in 12 patients with RAG mutations presenting with Omenn syndrome (n = 5), leaky SCID (n = 3), or CID-G/AI (n = 4). Restriction of repertoire diversity skewed usage of variable (V), diversity (D), and joining (J) segment genes, and abnormalities of CDR3 length distribution were progressively more prominent in patients with a more severe phenotype. Skewed usage of V, D, and J segment genes was present also within unique sequences, indicating a primary restriction of repertoire. Patients with Omenn syndrome had a high proportion of class-switched immunoglobulin heavy chain transcripts and increased somatic hypermutation rate, suggesting in vivo activation of these B cells. These data provide a framework to better understand the phenotypic heterogeneity of RAG deficiency.


Journal of Clinical Immunology | 2016

Ligase-4 Deficiency Causes Distinctive Immune Abnormalities in Asymptomatic Individuals

Kerstin Felgentreff; Sachin N. Baxi; Yu Nee Lee; Kerry Dobbs; Lauren A. Henderson; Krisztian Csomos; Erdyni Tsitsikov; Mary Armanios; Jolan E. Walter; Luigi D. Notarangelo

PurposeDNA Ligase 4 (LIG4) is a key factor in the non-homologous end-joining (NHEJ) DNA double-strand break repair pathway needed for V(D)J recombination and the generation of the T cell receptor and immunoglobulin molecules. Defects in LIG4 result in a variable syndrome of growth retardation, pancytopenia, combined immunodeficiency, cellular radiosensitivity, and developmental delay.MethodsWe diagnosed a patient with LIG4 syndrome by radiosensitivity testing on peripheral blood cells, and established that two of her four healthy siblings carried the same compound heterozygous LIG4 mutations. An extensive analysis of the immune phenotype, cellular radiosensitivity, telomere length, and T and B cell antigen receptor repertoire was performed in all siblings.ResultsIn the three genotypically affected individuals, variable severities of radiosensitivity, alterations of T and B cell counts with an increased percentage of memory cells, and hypogammaglobulinemia, were noticed. Analysis of T and B cell antigen receptor repertoires demonstrated increased usage of alternative microhomology-mediated end-joining (MHMEJ) repair, leading to diminished N nucleotide addition and shorter CDR3 length. However, overall repertoire diversity was preserved.ConclusionsWe demonstrate that LIG4 syndrome presents with high clinical variability even within the same family, and that distinctive immunologic abnormalities may be observed also in yet asymptomatic individuals.


Frontiers in Immunology | 2017

Natural killer cells from patients with recombinase-activating gene and non-homologous end joining gene defects comprise a higher frequency of CD56bright NKG2A+++ cells, and yet display increased degranulation and higher perforin content

Kerry Dobbs; Giovanna Tabellini; Enrica Calzoni; Ornella Patrizi; Paula Martinez; Silvia Giliani; Daniele Moratto; Waleed Al-Herz; Caterina Cancrini; Morton J. Cowan; Jacob Bleesing; Claire Booth; David Buchbinder; Siobhan O. Burns; Talal A. Chatila; Janet Chou; Vanessa Daza-Cajigal; Lisa Ott de Bruin; Maite de la Morena; Gigliola Di Matteo; Andrea Finocchi; Raif S. Geha; Rakesh K. Goyal; Anthony Hayward; Steven M. Holland; Chiung Hui Huang; Maria Kanariou; Alejandra King; Blanka M. Kaplan; Anastasiya Kleva

Mutations of the recombinase-activating genes 1 and 2 (RAG1 and RAG2) in humans are associated with a broad range of phenotypes. For patients with severe clinical presentation, hematopoietic stem cell transplantation (HSCT) represents the only curative treatment; however, high rates of graft failure and incomplete immune reconstitution have been observed, especially after unconditioned haploidentical transplantation. Studies in mice have shown that Rag−/− natural killer (NK) cells have a mature phenotype, reduced fitness, and increased cytotoxicity. We aimed to analyze NK cell phenotype and function in patients with mutations in RAG and in non-homologous end joining (NHEJ) genes. Here, we provide evidence that NK cells from these patients have an immature phenotype, with significant expansion of CD56bright CD16−/int CD57− cells, yet increased degranulation and high perforin content. Correlation was observed between in vitro recombinase activity of the mutant proteins, NK cell abnormalities, and in vivo clinical phenotype. Addition of serotherapy in the conditioning regimen, with the aim of depleting the autologous NK cell compartment, may be important to facilitate engraftment and immune reconstitution in patients with RAG and NHEJ defects treated by HSCT.

Collaboration


Dive into the Kerry Dobbs's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sung-Yun Pai

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Yu Nee Lee

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Caterina Cancrini

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Stefano Volpi

Istituto Giannina Gaslini

View shared research outputs
Researchain Logo
Decentralizing Knowledge