Network


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

Hotspot


Dive into the research topics where Ivan K. Chinn is active.

Publication


Featured researches published by Ivan K. Chinn.


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.


The Journal of Allergy and Clinical Immunology | 2017

Update on the use of immunoglobulin in human disease: A review of evidence.

Elena E. Perez; Jordan S. Orange; Francisco A. Bonilla; Javier Chinen; Ivan K. Chinn; Morna J. Dorsey; Yehia M. El-Gamal; Terry Harville; Elham Hossny; Bruce Mazer; Robert P. Nelson; Elizabeth Secord; Stanley C. Jordan; E. Richard Stiehm; Ashley Vo; Mark Ballow

&NA; Human immunoglobulin preparations for intravenous or subcutaneous administration are the cornerstone of treatment in patients with primary immunodeficiency diseases affecting the humoral immune system. Intravenous preparations have a number of important uses in the treatment of other diseases in humans as well, some for which acceptable treatment alternatives do not exist. We provide an update of the evidence‐based guideline on immunoglobulin therapy, last published in 2006. Given the potential risks and inherent scarcity of human immunoglobulin, careful consideration of its indications and administration is warranted.


Immunology and Allergy Clinics of North America | 2015

Severe Combined Immunodeficiency Disorders

Ivan K. Chinn; William T. Shearer

Severe combined immunodeficiency disorders represent pediatric emergencies due to absence of adaptive immune responses to infections. The conditions result from either intrinsic defects in T-cell development (ie, severe combined immunodeficiency disease [SCID]) or congenital athymia (eg, complete DiGeorge anomaly). Hematopoietic stem cell transplant provides the only clinically approved cure for SCID, although gene therapy research trials are showing significant promise. For greatest survival, patients should undergo transplant before 3.5 months of age and before the onset of infections. Newborn screening programs have yielded successful early identification and treatment of infants with SCID and congenital athymia in the United States.


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.


Journal of Clinical Investigation | 2017

Biallelic mutations in IRF8 impair human NK cell maturation and function

Emily M. Mace; Venetia Bigley; Justin T. Gunesch; Ivan K. Chinn; Laura S. Angelo; Matthew A. Care; Sheetal Maisuria; Michael Keller; Sumihito Togi; Levi B. Watkin; David F. LaRosa; Shalini N. Jhangiani; Donna M. Muzny; Asbjørg Stray-Pedersen; Zeynep Coban Akdemir; Jansen B. Smith; Mayra Hernández-Sanabria; Duy T. Le; Graham D. Hogg; Tram N. Cao; Aharon G. Freud; Eva P. Szymanski; Sinisa Savic; Matthew Collin; Andrew J. Cant; Richard A. Gibbs; Steven M. Holland; Michael A. Caligiuri; Keiko Ozato; Silke Paust

Human NK cell deficiencies are rare yet result in severe and often fatal disease, particularly as a result of viral susceptibility. NK cells develop from hematopoietic stem cells, and few monogenic errors that specifically interrupt NK cell development have been reported. Here we have described biallelic mutations in IRF8, which encodes an interferon regulatory factor, as a cause of familial NK cell deficiency that results in fatal and severe viral disease. Compound heterozygous or homozygous mutations in IRF8 in 3 unrelated families resulted in a paucity of mature CD56dim NK cells and an increase in the frequency of the immature CD56bright NK cells, and this impairment in terminal maturation was also observed in Irf8–/–, but not Irf8+/–, mice. We then determined that impaired maturation was NK cell intrinsic, and gene expression analysis of human NK cell developmental subsets showed that multiple genes were dysregulated by IRF8 mutation. The phenotype was accompanied by deficient NK cell function and was stable over time. Together, these data indicate that human NK cells require IRF8 for development and functional maturation and that dysregulation of this function results in severe human disease, thereby emphasizing a critical role for NK cells in human antiviral defense.


Journal of Clinical Investigation | 2016

Destabilized SMC5/6 complex leads to chromosome breakage syndrome with severe lung disease

Saskia N. van der Crabben; Marije P Hennus; Grant A. McGregor; Deborah I. Ritter; Sandesh C.S. Nagamani; Owen S. Wells; Magdalena Harakalova; Ivan K. Chinn; Aaron Alt; Lucie Vondrová; Ron Hochstenbach; Joris M. van Montfrans; Suzanne W. J. Terheggen-Lagro; Stef van Lieshout; Markus J. van Roosmalen; Ivo Renkens; Karen Duran; Isaac J. Nijman; Wigard P. Kloosterman; Eric A.M. Hennekam; Jordan S. Orange; Peter M. van Hasselt; David A. Wheeler; Jan Paleček; Alan R. Lehmann; Antony W. Oliver; Laurence H. Pearl; Sharon E. Plon; Johanne M. Murray; Gijs van Haaften

The structural maintenance of chromosomes (SMC) family of proteins supports mitotic proliferation, meiosis, and DNA repair to control genomic stability. Impairments in chromosome maintenance are linked to rare chromosome breakage disorders. Here, we have identified a chromosome breakage syndrome associated with severe lung disease in early childhood. Four children from two unrelated kindreds died of severe pulmonary disease during infancy following viral pneumonia with evidence of combined T and B cell immunodeficiency. Whole exome sequencing revealed biallelic missense mutations in the NSMCE3 (also known as NDNL2) gene, which encodes a subunit of the SMC5/6 complex that is essential for DNA damage response and chromosome segregation. The NSMCE3 mutations disrupted interactions within the SMC5/6 complex, leading to destabilization of the complex. Patient cells showed chromosome rearrangements, micronuclei, sensitivity to replication stress and DNA damage, and defective homologous recombination. This work associates missense mutations in NSMCE3 with an autosomal recessive chromosome breakage syndrome that leads to defective T and B cell function and acute respiratory distress syndrome in early childhood.


Frontiers in Pediatrics | 2017

First Case of CD40LG Deficiency in Ecuador, Diagnosed after Whole Exome Sequencing in a Patient with Severe Cutaneous Histoplasmosis

Luis Alberto Pedroza; Nina Guerrero; Asbjørg Stray-Pedersen; Cristina Tafur; Roque Macias; Greta Muñoz; Zeynep Coban Akdemir; Shalini N. Jhangiani; Levi B. Watkin; Ivan K. Chinn; James R. Lupski; Jordan S. Orange

Severe infections with Histoplasma capsulatum are commonly observed in patient with secondary immunodeficiency disorders. We report a two and a half years old boy previously healthy with disseminated cutaneous histoplasmosis. Using whole exome sequencing, we found an indel mutation at the CD40LG gene, suggesting a diagnosis of hyper-IgM (HIGM) syndrome, even in the absence of the usual features for the disease. Interestingly, the patient lives in a region endemic for histoplasmosis. The unusual infections in our case suggest that in children with severe histoplasmosis and resident in endemic areas, HIGM syndrome should be considered as a diagnosis.


Blood | 2018

Genetic and mechanistic diversity in pediatric hemophagocytic lymphohistiocytosis

Ivan K. Chinn; Olive S. Eckstein; Erin C. Peckham-Gregory; Baruch R. Goldberg; Lisa R. Forbes; Sarah K. Nicholas; Emily M. Mace; Tiphanie P. Vogel; Harshal Abhyankar; Maria I. Diaz; Helen E. Heslop; Robert A. Krance; Caridad Martinez; Trung C. Nguyen; Dalia A. Bashir; Jordana Goldman; Asbjørg Stray-Pedersen; Luis Alberto Pedroza; M. Cecilia Poli; Juan C. Aldave-Becerra; Sean A. McGhee; Waleed Al-Herz; Aghiad Chamdin; Zeynep H. Coban-Akdemir; Shalini N. Jhangiani; Donna M. Muzny; Tram N. Cao; Diana N. Hong; Richard A. Gibbs; James R. Lupski

The HLH-2004 criteria are used to diagnose hemophagocytic lymphohistiocytosis (HLH), yet concern exists for their misapplication, resulting in suboptimal treatment of some patients. We sought to define the genomic spectrum and associated outcomes of a diverse cohort of children who met the HLH-2004 criteria. Genetic testing was performed clinically or through research-based whole-exome sequencing. Clinical metrics were analyzed with respect to genomic results. Of 122 subjects enrolled over the course of 17 years, 101 subjects received genetic testing. Biallelic familial HLH (fHLH) gene defects were identified in only 19 (19%) and correlated with presentation at younger than 1 year of age (P < .0001). Digenic fHLH variants were observed but lacked statistical support for disease association. In 28 (58%) of 48 subjects, research whole-exome sequencing analyses successfully identified likely molecular explanations, including underlying primary immunodeficiency diseases, dysregulated immune activation and proliferation disorders, and potentially novel genetic conditions. Two-thirds of patients identified by the HLH-2004 criteria had underlying etiologies for HLH, including genetic defects, autoimmunity, and malignancy. Overall survival was 45%, and increased mortality correlated with HLH triggered by infection or malignancy (P < .05). Differences in survival did not correlate with genetic profile or extent of therapy. HLH should be conceptualized as a phenotype of critical illness characterized by toxic activation of immune cells from different underlying mechanisms. In most patients with HLH, targeted sequencing of fHLH genes remains insufficient for identifying pathogenic mechanisms. Whole-exome sequencing, however, may identify specific therapeutic opportunities and affect hematopoietic stem cell transplantation options for these patients.


American Journal of Human Genetics | 2018

Identifying Genes Whose Mutant Transcripts Cause Dominant Disease Traits by Potential Gain-of-Function Alleles

Zeynep Coban-Akdemir; Janson J. White; Xiaofei Song; Shalini N. Jhangiani; Jawid M. Fatih; Tomasz Gambin; Yavuz Bayram; Ivan K. Chinn; Ender Karaca; Jaya Punetha; Cecilia Poli; Eric Boerwinkle; Chad A. Shaw; Jordan S. Orange; Richard A. Gibbs; Tuuli Lappalainen; James R. Lupski; Claudia M.B. Carvalho

Premature termination codon (PTC)-bearing transcripts are often degraded by nonsense-mediated decay (NMD) resulting in loss-of-function (LoF) alleles. However, not all PTCs result in LoF mutations, i.e., some such transcripts escape NMD and are translated to truncated peptide products that result in disease due to gain-of-function (GoF) effects. Since the location of the PTC is a major factor determining transcript fate, we hypothesized that depletion of protein-truncating variants (PTVs) within the gene region predicted to escape NMD in control databases could provide a rank for genic susceptibility for disease through GoF versus LoF. We developed an NMD escape intolerance score to rank genes based on the depletion of PTVs that would render them able to escape NMD using the Atherosclerosis Risk in Communities Study (ARIC) and the Exome Aggregation Consortium (ExAC) control databases, which was further used to screen the Baylor-Center for Mendelian Genomics disease database. This analysis revealed 1,996 genes significantly depleted for PTVs that are predicted to escape from NMD, i.e., PTVesc; further studies provided evidence that revealed a subset as candidate genes underlying Mendelian phenotypes. Importantly, these genes have characteristically low pLI scores, which can cause them to be overlooked as candidates for dominant diseases. Collectively, we demonstrate that this NMD escape intolerance score is an effective and efficient tool for gene discovery in Mendelian diseases due to production of truncated or altered proteins. More importantly, we provide a complementary analytical tool to aid identification of genes associated with dominant traits through a mechanism distinct from LoF.


bioRxiv | 2017

Ruxolitinib partially reverses functional NK cell deficiency in patients with STAT1 gain-of-function mutations

Alexander Vargas Hernandez; Emily M. Mace; Ofer Zimmerman; Christa S. Zerbe; Alexandra F. Freeman; Sergio D. Rosenzweig; Jennifer W. Leiding; Troy R. Torgerson; Matthew C. Altman; Edith Schussler; Charlotte Cunningham-Rundles; Ivan K. Chinn; Imelda C. Hanson; Nicholas L. Rider; Steven M. Holland; Jordan S. Orange; Lisa R. Forbes

Background Natural Killer (NK) cells are critical innate effector cells whose development is dependent on the JAK-STAT pathway. NK deficiency can result in severe or refractory viral infections. Patients with Signal Transducer and Activator of Transcription (STAT)1 gain of function (GOF) mutations have increased viral susceptibility. Objective We sought to investigate NK cell function in STAT1 GOF patients. Methods: NK cell phenotype and function were determined in 16 STAT1 GOF patients. Methods NK cell phenotype and function were determined in 16 STAT1 GOF patients.NK cell lines expressing patient mutations were generated with CRISPR-Cas9 mediated gene editing. STAT1 GOF NK cells were treated in vitro with ruxolitinib. Results Peripheral blood NK cells from of STAT1 GOF patients had impaired terminal maturation. Specifically, patients with STAT1 GOF mutations have immature CD56dim NK cells with decreased expression of CD16, perforin, CD57 and impaired cytolytic function. STAT1 phosphorylation was elevated but STAT5 was aberrantly phosphorylated in response to IL-2 stimulation. Upstream inhibition of STAT signaling with the small molecule JAK1/2 inhibitor ruxolitinib in vitro and in vivo restored perforin expression in CD56dim NK cells and partially restored NK cell cytotoxic function. Conclusions Properly regulated STAT1 signaling is critical for NK cell maturation and function. Modulation of elevated STAT1 phosphorylation with ruxolitinib is an important option for therapeutic intervention in patients with STAT1 GOF mutations.

Collaboration


Dive into the Ivan K. Chinn's collaboration.

Top Co-Authors

Avatar

Jordan S. Orange

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Lisa R. Forbes

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Emily M. Mace

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

James R. Lupski

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
Top Co-Authors

Avatar

Richard A. Gibbs

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donna M. Muzny

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge