Network


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

Hotspot


Dive into the research topics where Huie Jing is active.

Publication


Featured researches published by Huie Jing.


The New England Journal of Medicine | 2009

Combined Immunodeficiency Associated with DOCK8 Mutations

Qian Zhang; Jeremiah C. Davis; Ian T. Lamborn; Alexandra F. Freeman; Huie Jing; Amanda J. Favreau; Helen F. Matthews; Joie Davis; Maria L. Turner; Gulbu Uzel; Steven M. Holland; Helen C. Su

BACKGROUND Recurrent sinopulmonary and cutaneous viral infections with elevated serum levels of IgE are features of some variants of combined immunodeficiency. The genetic causes of these variants are unknown. METHODS We collected longitudinal clinical data on 11 patients from eight families who had recurrent sinopulmonary and cutaneous viral infections. We performed comparative genomic hybridization arrays and targeted gene sequencing. Variants with predicted loss-of-expression mutations were confirmed by means of a quantitative reverse-transcriptase-polymerase-chain-reaction assay and immunoblotting. We evaluated the number and function of lymphocytes with the use of in vitro assays and flow cytometry. RESULTS Patients had recurrent otitis media, sinusitis, and pneumonias; recurrent Staphylococcus aureus skin infections with otitis externa; recurrent, severe herpes simplex virus or herpes zoster infections; extensive and persistent infections with molluscum contagiosum; and human papillomavirus infections. Most patients had severe atopy with anaphylaxis; several had squamous-cell carcinomas, and one had T-cell lymphoma-leukemia. Elevated serum IgE levels, hypereosinophilia, low numbers of T cells and B cells, low serum IgM levels, and variable IgG antibody responses were common. Expansion in vitro of activated CD8 T cells was impaired. Novel homozygous or compound heterozygous deletions and point mutations in the gene encoding the dedicator of cytokinesis 8 protein (DOCK8) led to the absence of DOCK8 protein in lymphocytes. CONCLUSIONS Autosomal recessive DOCK8 deficiency is associated with a novel variant of combined immunodeficiency.


Journal of Experimental Medicine | 2012

Congenital B cell lymphocytosis explained by novel germline CARD11 mutations

Andrew L. Snow; Wenming Xiao; Jeffrey R. Stinson; Wei Lu; Benjamin Chaigne-Delalande; Lixin Zheng; Stefania Pittaluga; Helen F. Matthews; Roland Schmitz; Sameer Jhavar; Stefan Kuchen; Lela Kardava; Wei Wang; Ian T. Lamborn; Huie Jing; Mark Raffeld; Susan Moir; Thomas A. Fleisher; Louis M. Staudt; Helen C. Su; Michael J. Lenardo

Germline mutations in CARD11 that result in constitutive NF-κB activation and selective B cell expansion underlie congenital B cell lymphocytosis.


Archives of Dermatology | 2012

Cutaneous Manifestations of DOCK8 Deficiency Syndrome

Emily Y. Chu; Alexandra F. Freeman; Huie Jing; Edward W. Cowen; Joie Davis; Helen C. Su; Steven M. Holland; Maria L. Turner

BACKGROUND Mutations in the dedicator of cytokinesis 8 gene (DOCK8) cause a combined primary immunodeficiency syndrome that is characterized by elevated serum IgE levels, depressed IgM levels, eosinophilia, sinopulmonary infections, cutaneous viral infections, and lymphopenia. Many patients with DOCK8 deficiency were previously thought to have a variant of Jobs syndrome. Distinguishing between DOCK8 deficiency and Jobs syndrome, also referred to as autosomal dominant hyper-IgE syndrome, on the basis of clinical findings alone is challenging. The discovery of the DOCK8 mutation has made it possible to differentiate the cutaneous manifestations of these hyper-IgE syndromes. OBSERVATIONS Twenty-one patients from 14 families with confirmed homozygous or compound heterozygous mutations in DOCK8 were evaluated. Clinical findings included dermatitis, asthma, food and environmental allergies, recurrent sinopulmonary infections, staphylococcal skin abscesses, and severe cutaneous viral infections. Malignant neoplasms, including aggressive cutaneous T-cell lymphoma, anal and vulvar squamous cell carcinomas, and diffuse large B-cell lymphoma, developed in 5 patients during adolescence and young adulthood. CONCLUSIONS DOCK8 deficiency and Jobs syndrome share several clinical features, including elevated serum IgE levels, dermatitis, recurrent sinopulmonary infections, and cutaneous staphylococcal abscesses. However, the presence of recalcitrant, widespread cutaneous viral infections, asthma, and food and environmental allergies, as well as the absence of newborn rash and coarse facies, favors the clinical diagnosis of DOCK8 deficiency. Rates of malignancy and overall mortality in patients with DOCK8 deficiency were higher than in those with Jobs syndrome, highlighting the value of distinguishing between these conditions and the importance of close monitoring for neoplasia.


Journal of Clinical Immunology | 2012

Additional Diverse Findings Expand the Clinical Presentation of DOCK8 Deficiency

Ozden Sanal; Huie Jing; Tuba Turul Ozgur; Deniz Cagdas Ayvaz; Dara M. Strauss-Albee; Sibel Ersoy-Evans; Ilhan Tezcan; Gulten Turkkani; Helen F. Matthews; Goknur Haliloglu; Aysel Yüce; Bilgehan Yalçın; Özay Gököz; Kader Karli Oguz; Helen C. Su

We describe seven Turkish children with DOCK8 deficiency who have not been previously reported. Three patients presented with typical features of recurrent or severe cutaneous viral infections, atopic dermatitis, and recurrent respiratory or gastrointestinal tract infections. However, four patients presented with other features. Patient 1–1 featured sclerosing cholangitis and colitis; patient 2–1, granulomatous soft tissue lesion and central nervous system involvement, with primary central nervous system lymphoma found on follow-up; patient 3–1, a fatal metastatic leiomyosarcoma; and patient 4–2 showed no other symptoms initially besides atopic dermatitis. Similar to other previously reported Turkish patients, but in contrast to patients of non-Turkish ethnicity, the patients’ lymphopenia was primarily restricted to CD4+ T cells. Patients had homozygous mutations in DOCK8 that altered splicing, introduced premature terminations, destabilized protein, or involved large deletions within the gene. Genotyping of remaining family members showed that DOCK8 deficiency is a fully penetrant, autosomal recessive disease. In our patients, bone marrow transplantation resulted in rapid improvement followed by disappearance of viral skin lesions, including lesions resembling epidermodysplasia verruciformis, atopic dermatitis, and recurrent infections. Particularly for patients who feature unusual clinical manifestations, immunological testing, in conjunction with genetic testing, can prove invaluable in diagnosing DOCK8 deficiency and providing potentially curative treatment.


Annals of the New York Academy of Sciences | 2011

DOCK8 deficiency: DOCK8 deficiency

Helen C. Su; Huie Jing; Qian Zhang

The discovery that loss‐of‐function mutations in the gene DOCK8 are responsible for most forms of autosomal recessive hyper‐IgE syndrome and some forms of combined immunodeficiency without elevated serum IgE has led to studies into the immunopathogenesis of this disease. In this review, we relate the clinical features of this disease to studies using patients’ cells and a mouse model of Dock8 deficiency, which have revealed how DOCK8 regulates T and B cell numbers and functions. The results of these studies help to explain how the absence of DOCK8 contributes to patients’ susceptibility to viral, fungal, and bacterial infections. However, unanswered questions remain regarding how the absence of DOCK8 also leads to high IgE and allergic disease, predisposition for malignancy, and unusual clinical features, such as CNS abnormalities and autoimmunity, observed in some patients.


The Journal of Allergy and Clinical Immunology | 2014

Somatic reversion in dedicator of cytokinesis 8 immunodeficiency modulates disease phenotype

Huie Jing; Qian Zhang; Yu Zhang; Brenna J. Hill; Christopher G. Dove; Erwin W. Gelfand; T. Prescott Atkinson; Gulbu Uzel; Helen F. Matthews; Peter Mustillo; David B. Lewis; Fotini Kavadas; I. Celine Hanson; Ashish Kumar; Raif S. Geha; Steven M. Holland; Alexandra F. Freeman; Helen C. Su

BACKGROUND Autosomal recessive loss-of-function mutations in dedicator of cytokinesis 8 (DOCK8) cause a combined immunodeficiency characterized by atopy, recurrent infections, and cancer susceptibility. A genotype-phenotype explanation for the variable disease expression is lacking. OBJECTIVE We investigated whether reversions contributed to the variable disease expression. METHODS Patients followed at the National Institutes of Healths Clinical Center were studied. We performed detailed genetic analyses and intracellular flow cytometry to detect DOCK8 protein expression within lymphocyte subsets. RESULTS We identified 17 of 34 DOCK8-deficient patients who had germline mutations with variable degrees of reversion caused by somatic repair. Somatic repair of the DOCK8 mutations resulted from second-site mutation, original-site mutation, gene conversion, and intragenic crossover. Higher degrees of reversion were associated with recombination-mediated repair. DOCK8 expression was restored primarily within antigen-experienced T cells or natural killer cells but less so in naive T or B cells. Several patients exhibited multiple different repair events. Patients who had reversions were older and had less severe allergic disease, although infection susceptibility persisted. No patients were cured without hematopoietic cell transplantation. CONCLUSIONS In patients with DOCK8 deficiency, only certain combinations of germline mutations supported secondary somatic repair. Those patients had an ameliorated disease course with longer survival but still had fatal complications or required hematopoietic cell transplantation. These observations support the concept that some DOCK8-immunodeficient patients have mutable mosaic genomes that can modulate disease phenotype over time.


Journal of Experimental Medicine | 2017

Combined immunodeficiency and Epstein-Barr virus?induced B cell malignancy in humans with inherited CD70 deficiency

Hassan Abolhassani; Aydan Ikinciogullari; Huie Jing; Stephan Borte; Marcus Buggert; Likun Du; Mami Matsuda-Lennikov; Rosa Romano; Rozina Caridha; Sangeeta Bade; Yu Zhang; Juliet Wairimu Frederiksen; Mingyan Fang; Sevgi Köstel Bal; Sule Haskologlu; Figen Dogu; Nurdan Tacyildiz; Helen F. Matthews; Joshua McElwee; Emma Gostick; David A. Price; Umaimainthan Palendira; Asghar Aghamohammadi; Bertrand Boisson; Nima Rezaei; Annika C. Karlsson; Michael J. Lenardo; Jean-Laurent Casanova; Lennart Hammarström; Stuart G. Tangye

In this study, we describe four patients from two unrelated families of different ethnicities with a primary immunodeficiency, predominantly manifesting as susceptibility to Epstein-Barr virus (EBV)–related diseases. Three patients presented with EBV-associated Hodgkin’s lymphoma and hypogammaglobulinemia; one also had severe varicella infection. The fourth had viral encephalitis during infancy. Homozygous frameshift or in-frame deletions in CD70 in these patients abolished either CD70 surface expression or binding to its cognate receptor CD27. Blood lymphocyte numbers were normal, but the proportions of memory B cells and EBV-specific effector memory CD8+ T cells were reduced. Furthermore, although T cell proliferation was normal, in vitro–generated EBV-specific cytotoxic T cell activity was reduced because of CD70 deficiency. This reflected impaired activation by, rather than effects during killing of, EBV-transformed B cells. Notably, expression of 2B4 and NKG2D, receptors implicated in controlling EBV infection, on memory CD8+ T cells from CD70-deficient individuals was reduced, consistent with their impaired killing of EBV-infected cells. Thus, autosomal recessive CD70 deficiency is a novel cause of combined immunodeficiency and EBV-associated diseases, reminiscent of inherited CD27 deficiency. Overall, human CD70–CD27 interactions therefore play a nonredundant role in T and B cell–mediated immunity, especially for protection against EBV and humoral immunity.


The Journal of Allergy and Clinical Immunology | 2014

Vaccine strain varicella-zoster virus-induced central nervous system vasculopathy as the presenting feature of DOCK8 deficiency.

Angela Sabry; Pia J. Hauk; Huie Jing; Helen C. Su; Nicholas V. Stence; David M. Mirsky; Maria A. Nagel; Jordan K. Abbott; Leonard L. Dragone; Jennifer Armstrong-Wells; Donna Curtis; Randall J. Cohrs; D. Scott Schmid; Donald H. Gilden; Erwin W. Gelfand

To the Editor: In contrast to autosomal dominant forms of hyper-IgE syndrome resulting from mutations in the STAT3 gene, autosomal recessive dedicator of cytokinesis 8 (DOCK8) deficiency, results in susceptibility to cutaneous viral infections, eosinophilia, and allergic disease. CNS manifestations have been reported in patients with DOCK8 deficiency, but progressive multifocal leukoencephalopathy caused by JC virus has been considered the only known viral etiology to date (1, 2). A 6 year-old boy with atopy and recurrent peripheral blood eosinophilia developed acute intermittent vomiting, diarrhea, headache, and dizziness. Magnetic resonance imaging (MRI) of the brain was normal. One day later, he began giggling inappropriately, experienced left leg paresthesias, urinary incontinence, and fell upon attempting to stand. Repeat MRI with axial diffusion-weighted images revealed multiple areas of acute infarction in areas supplied by both anterior cerebral arteries, a branch of the anterior cerebral artery, the right posterior cerebral artery, and the left middle cerebral artery (Figure 1A). A complete blood count revealed a total eosinophil count of 2,160/μL, but no other abnormalities. He was treated with acetylsalicylic acid and intravenous methylprednisolone followed by oral prednisolone, 2 mg/kg for 3 days after which prednisolone was maintained at 1 mg/kg daily. Figure 1 Manifestation of vaccine strain varicella zoster virus-induced central nervous system vasculopathy On presentation to us one month later, he had developed paresthesias of his hands. Neurological examination showed only mild weakness in the hands. Computed tomography angiogram revealed diffuse vasculopathy. Subsequent 3-dimensional time of flight magnetic resonance angiographic imaging of the circle of Willis revealed vascular narrowing and post-stenotic dilatation (Figure 1B). Post-contrast T1W black blood arterial wall imaging illustrated avid enhancement and thickening of the distal supraclinoid internal carotid arterial walls bilaterally (Figure 1C). Cerebral spinal fluid (CSF) was obtained. While awaiting results of testing for viral CNS infections, the patient was treated with acyclovir, 30 mg/kg intravenous daily, and maintained on oral prednisolone, 2 mg/kg/day. The CSF at the time of diagnosis of vasculopathy contained 21 mononuclear cells and no red blood cells; protein was 39 mg/dL and glucose was 72 mg/dL. Quantitative PCR (Focus Diagnostics Reference Laboratory, Cypress, CA) amplified 7,116 copies of VZV DNA/mL in the CSF. To determine the VZV genotype, Forster Resonance Energy Transfer PCR was used to identify specific VZV DNA sequence polymorphisms within VZV open reading frames 38, 54, and 62, which distinguishes vaccine VZV from wild-type virus (Online Supplement Table 1) (3). This confirmed Oka varicella vaccine strain in the CSF. The CSF also contained anti-VZV IgG antibodies, but no antibodies to HSV-1 or HSV-2, enterovirus, cytomegalovirus, or Epstein-Barr virus were detected and the respective PCR analysis were similarly negative. The serum to CSF ratio of anti-VZV IgG antibody was markedly decreased (ratio 2.3) compared to albumin (ratio 149). Two weeks after treatment with acyclovir, the neurologic examination was completely normal, the CSF was acellular, and PCR was negative for VZV DNA. The patient received routine immunizations including VZV vaccination at 12 months of age. He had 3 episodes of vesicular rashes at ages 3, 4, and 5 years, which always occurred after completing a course of oral corticosteroids. The first rash tested positive for HSV. The second rash was identical. The third rash at age 5 years with vesicles on the lower thigh was diagnosed clinically as zoster. All rashes resolved on oral acyclovir. Past history included early-onset atopic dermatitis, food allergies often with anaphylaxis, biopsy-confirmed eosinophilic esophagitis, asthma and recurrent upper respiratory tract infections. Peripheral blood eosinophilia peaked at 9,000 eosinophils/μL and serum IgE at 472 IU/mL. During flares of respiratory or skin disease, he was treated with oral corticosteroids intermittently for 3 to 5 days, a few times annually. The frequency of such treatment increased and between ages 5½ and 6, he was treated with oral corticosteroids 1 to 2 times monthly during the prior 6 months. Conventional comparative genomic hybridization array analysis revealed a large deletion of exons 1 to 13 in a single allele of the DOCK8 gene. PCR analysis of genomic DNA and DOCK8 gene sequencing identified a single base pair mutation on the opposite allele at exon 12, resulting in a frame shift and premature stop codon: c.1266delC, p.Y423TfsX18, based upon reference sequence NM_00193536.1, isoform 3 (Figure 2A). Western Blot analysis confirmed lack of DOCK8 protein expression (Figure 2B). Parental testing demonstrated that the large exon deletion was inherited from the mother while the point mutation was inherited from the father. Immunological findings are summarized in Supplemental Table 2. Figure 2 DOCK8 molecular analyses The neurological symptoms and signs, imaging and CSF abnormalities, virological studies, and response to antiviral treatment were all features characteristically seen in VZV vasculopathy (4). Evidence of both focal and diffuse CNS disease was corroborated by widespread infarction produced by stenosis of multiple large cerebral arteries. The CSF examination revealed a pleocytosis as found in two-thirds of patients with VZV vasculopathy and both VZV DNA and anti-VZV IgG antibody with reduced serum/CSF ratios of anti-VZV IgG antibody were detected, indicative of intrathecal synthesis of anti-VZV IgG. Analysis of VZV DNA in CSF revealed that the VZV genotype was vaccine strain, demonstrating, for the first time, that VZV reactivation after vaccination in childhood can result in VZV vasculopathy. Although the underlying immunological consequences of DOCK8 deficiency remain to be fully elucidated, multiple immune system abnormalities may account for enhanced susceptibility to viral infection including impaired dendritic cell migration affecting T cell priming, lymphopenia, defective CD8 T cell activation and expansion, decreased production of the anti-viral cytokines IFN-γ and TNF-α, impaired T cell survival, decreased NK cell cytotoxicity, and antibody abnormalities (5-9). Germinal center formation and survival of germinal center B cells are impaired in DOCK8 deficiency, leading to defective long-lived antibody production (8). Responses to protein or polysaccharide-conjugated vaccines are often variable while responses to previously encountered viruses such as HSV and VZV have been normal as demonstrated here for VZV antibodies in serum and CSF. As in some other young patients with DOCK8 deficiency, this patient tended toward the milder spectrum of the disease with a relatively limited history of cutaneous infections, absence of severe systemic infections other than chronic mild otitis, and the modestly elevated serum IgE level. Instead, eosinophilia and moderate-severe eczema, asthma, and food sensitivities predominated. To control these conditions, courses of oral corticosteroids were administered with increased frequency over time. Interestingly, each of the 3 episodes of cutaneous viral infections with HSV or VZV was preceded by a course of oral corticosteroids. It is possible corticosteroids alone enabled the activation of vaccine strain VZV infection. More likely, the use of oral corticosteroids to gain disease control reduced the patient’s “immunologic threshold” and together resulted in reactivation of vaccine strain VZV and subsequent vasculopathy. In summary, a young patient with significant atopic disease and widespread infarction produced by stenosis of multiple large cerebral arteries was shown to express novel mutations on both alleles of the DOCK8 gene. For the first time, VZV vasculopathy was shown to be due to the vaccine strain. This case highlights the importance of considering the possibility of DOCK8 deficiency in the context of severe allergic disease and the potential risks for CNS infection including VZV vaccine-related vasculopathy.


Journal of Experimental Medicine | 2017

Recurrent rhinovirus infections in a child with inherited MDA5 deficiency

Ian T. Lamborn; Huie Jing; Yu Zhang; Scott Drutman; Jordan K. Abbott; Shirin Munir; Sangeeta Bade; Heardley M. Murdock; Celia Santos; Linda G. Brock; Evan Masutani; Emmanuel Y. Fordjour; Joshua McElwee; Jason D. Hughes; Dave P. Nichols; Aziz Belkadi; Andrew J. Oler; Corinne S. Happel; Helen F. Matthews; Laurent Abel; Peter L. Collins; Kanta Subbarao; Erwin W. Gelfand; Michael J. Ciancanelli; Jean-Laurent Casanova; Helen C.F. Su

MDA5 is a cytosolic sensor of double-stranded RNA (ds)RNA including viral byproducts and intermediates. We studied a child with life-threatening, recurrent respiratory tract infections, caused by viruses including human rhinovirus (HRV), influenza virus, and respiratory syncytial virus (RSV). We identified in her a homozygous missense mutation in IFIH1 that encodes MDA5. Mutant MDA5 was expressed but did not recognize the synthetic MDA5 agonist/(ds)RNA mimic polyinosinic-polycytidylic acid. When overexpressed, mutant MDA5 failed to drive luciferase activity from the IFNB1 promoter or promoters containing ISRE or NF-&kgr;B sequence motifs. In respiratory epithelial cells or fibroblasts, wild-type but not knockdown of MDA5 restricted HRV infection while increasing IFN-stimulated gene expression and IFN-&bgr;/&lgr;. However, wild-type MDA5 did not restrict influenza virus or RSV replication. Moreover, nasal epithelial cells from the patient, or fibroblasts gene-edited to express mutant MDA5, showed increased replication of HRV but not influenza or RSV. Thus, human MDA5 deficiency is a novel inborn error of innate and/or intrinsic immunity that causes impaired (ds)RNA sensing, reduced IFN induction, and susceptibility to the common cold virus.


Journal of Clinical Immunology | 2016

Recent Advances in DOCK8 Immunodeficiency Syndrome

Qian Zhang; Huie Jing; Helen C. Su

Since the discovery of the genetic basis of DOCK8 immunodeficiency syndrome (DIDS) in 2009, several hundred patients worldwide have been reported, validating and extending the initial clinical descriptions. Importantly, the beneficial role of hematopoietic stem cell transplantation for this disease has emerged, providing impetus for improved diagnosis. Additionally, several groups have further elucidated the biological functions of DOCK8 in the immune system that help explain disease pathogenesis. Here, we summarize these recent developments.

Collaboration


Dive into the Huie Jing's collaboration.

Top Co-Authors

Avatar

Helen C. Su

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Helen F. Matthews

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Qian Zhang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Alexandra F. Freeman

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Yu Zhang

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ian T. Lamborn

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Steven M. Holland

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Erwin W. Gelfand

University of Colorado Denver

View shared research outputs
Top Co-Authors

Avatar

Michael J. Lenardo

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge