Network


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

Hotspot


Dive into the research topics where Chi A. Ma is active.

Publication


Featured researches published by Chi A. Ma.


Journal of Clinical Investigation | 2006

Impaired regulation of NF-κB and increased susceptibility to colitis-associated tumorigenesis in CYLD-deficient mice

Jun Zhang; Brigid Stirling; Stephane T. Temmerman; Chi A. Ma; Ivan J. Fuss; Jonathan M.J. Derry; Ashish Jain

Cylindromatosis (CYLD) is a deubiquitinating enzyme that is altered in patients with familial cylindromatosis, a condition characterized by numerous benign adnexal tumors. However, the regulatory function of CYLD remains unsettled. Here we show that the development of B cells, T cells, and myeloid cells was unaffected in CYLD-deficient mice, but that the activation of these cells with mediators of innate and adaptive immunity resulted in enhanced NF-kappaB and JNK activity associated with increased TNF receptor-associated factor 2 (TRAF2) and NF-kappaB essential modulator (NEMO) ubiquitination. CYLD-deficient mice were more susceptible to induced colonic inflammation and showed a dramatic increase in the incidence of tumors compared with controls in a colitis-associated cancer model. These results suggest that CYLD limits inflammation and tumorigenesis by regulating ubiquitination in vivo.


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 | 2004

Specific NEMO mutations impair CD40-mediated c-Rel activation and B cell terminal differentiation

Ashish Jain; Chi A. Ma; Eduardo López-Granados; Gary D. Means; William Brady; Jordan S. Orange; Shuying Liu; Steven M. Holland; Jonathan M.J. Derry

Hypomorphic mutations in the zinc finger domain of NF-kappaB essential modulator (NEMO) cause X-linked hyper-IgM syndrome with ectodermal dysplasia (XHM-ED). Here we report that patient B cells are characterized by an absence of Ig somatic hypermutation (SHM) and defective class switch recombination (CSR) despite normal induction of activation-induced cytidine deaminase (AID) and Iepsilon-Cepsilon transcripts. This indicates that AID expression alone is insufficient to support neutralizing antibody responses. Furthermore, we show that patient B cells stimulated with CD40 ligand are impaired in both p65 and c-Rel activation, and whereas addition of IL-4 can enhance p65 activity, c-Rel activity remains deficient. This suggests that these NF-kappaB components have different activation requirements and that IL-4 can augment some but not all NEMO-dependent NF-kappaB signaling. Finally, using microarray analysis of patient B cells we identified downstream effects of impaired NF-kappaB activation and candidate factors that may be necessary for CSR and SHM in B cells.


Human Mutation | 2008

A Novel Mutation in NFKBIA/IKBA Results in a Degradation- Resistant N-Truncated Protein and Is Associated With Ectodermal Dysplasia With Immunodeficiency

Eduardo Lopez-Granados; Jeffrey E. Keenan; Matthew C. Kinney; Harvey L. Leo; Neal Jain; Chi A. Ma; Ralph Quinones; Erwin W. Gelfand; Ashish Jain

Alterations in nuclear factor kappa B (NF‐κB) essential modulator (NEMO; HUGO‐approved symbol IKBKG) underlie most cases of ectodermal dysplasia with immune deficiency (EDI), a human disorder characterized by anhidrosis with diminished immunity. EDI has also been associated with a single heterozygous mutation at position Ser32 of the NF‐κB inhibitor IκBα, one of two phosphorylation sites that are essential for targeting IκBα for proteasomal degradation and hence for activation of NF‐κB. We report a novel heterozygous nonsense mutation in the IKBA (HUGO‐approved symbol, NFKBIA) gene of a 1‐year‐old male child with EDI that introduces a premature termination codon at position Glu14. An in‐frame methionine downstream of the nonsense mutation allows for reinitiation of translation. The resulting N‐terminally truncated protein lacks both serine phosphorylation sites and inhibits NF‐κB signaling by functioning as a dominant negative on NF‐κB activity in lymphocytes and monocytes. These findings support the scanning model for translation initiation in eukaryotes and confirm the critical role of the NF‐κB in the human immune response. Hum Mutat 29(6), 861–868, 2008. Published 2008, Wiley‐Liss, Inc.


Journal of Biological Chemistry | 2011

Aurora B Interacts with NIR-p53, Leading to p53 Phosphorylation in Its DNA-binding Domain and Subsequent Functional Suppression

Liming Wu; Chi A. Ma; Yongge Zhao; Ashish Jain

NIR (novel INHAT repressor) is a transcriptional co-repressor with inhibitor of histone acetyltransferase (INHAT) activity and has previously been shown to physically interact with and suppress p53 transcriptional activity and function. However, the mechanism by which NIR suppresses p53 is not completely understood. Using a proteomic approach, we have identified the Aurora kinase B as a novel binding partner of NIR. We show that Aurora B, NIR and p53 exist in a protein complex in which Aurora B binds to NIR, thus also indirectly associates with p53. Functionally, overexpression of Aurora B or NIR suppresses p53 transcriptional activity, and depletion of Aurora B or NIR causes p53-dependent apoptosis and cell growth arrest, due to the up-regulation of p21 and Bax. We then demonstrate that Aurora B phosphorylates multiple sites in the p53 DNA-binding domain in vitro, and this phosphorylation probably also occurs in cells. Importantly, the Aurora B-mediated phosphorylation on Ser269 or Thr284 significantly compromises p53 transcriptional activity. Taken together, these results provide novel insight into NIR-mediated p53 suppression and also suggest an additional way for p53 regulation.


Journal of Biological Chemistry | 2011

The Deubiquitinase CYLD Targets Smad7 Protein to Regulate Transforming Growth Factor β (TGF-β) Signaling and the Development of Regulatory T Cells

Yongge Zhao; Angela M. Thornton; Matthew C. Kinney; Chi A. Ma; Jacob J. Spinner; Ivan J. Fuss; Ethan M. Shevach; Ashish Jain

Background: CYLD is a deubiquitinating enzyme (DUB) that hydrolyzes Lys-63-linked polyubiquitin chains that are attached covalently to cellular proteins. Results: CYLD knock-out mice have increased numbers of regulatory T cells (Tregs) in peripheral lymphoid organs but not in the thymus. Conclusion: CYLD regulates lysine 63-linked ubiquitination of Smad7 to control the development of peripheral Tregs. Significance: TGF-β signaling in T cells is regulated by lysine 63-Linked ubiquitination. CYLD is a lysine 63-deubiquitinating enzyme that inhibits NF-κB and JNK signaling. Here, we show that CYLD knock-out mice have markedly increased numbers of regulatory T cells (Tregs) in peripheral lymphoid organs but not in the thymus. In vitro stimulation of CYLD-deficient naive T cells with anti-CD3/28 in the presence of TGF-β led to a marked increase in the number of Foxp3-expressing T cells when compared with stimulated naive control CD4+ cells. Under endogenous conditions, CYLD formed a complex with Smad7 that facilitated CYLD deubiquitination of Smad7 at lysine 360 and 374 residues. Moreover, this site-specific ubiquitination of Smad7 was required for activation of TAK1 and p38 kinases. Finally, knockdown of Smad7 or inhibition of p38 activity in primary T cells impaired Treg differentiation. Together, our results show that CYLD regulates TGF-β signaling function in T cells and the development of Tregs through deubiquitination of Smad7.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Antibody deficiency associated with an inherited autosomal dominant mutation in TWEAK

Hong-Ying Wang; Chi A. Ma; Yongge Zhao; Xiying Fan; Qing Zhou; Pamela Edmonds; Gulbu Uzel; Joao Bosco Oliveira; Jordan S. Orange; Ashish Jain

Mutations in the TNF family of proteins have been associated with inherited forms of immune deficiency. Using an array-based sequencing assay, we identified an autosomal-dominant deficiency in TNF-like weak inducer of apoptosis (TWEAK; TNFSF12) in a kindred with recurrent infection and impaired antibody responses to protein and polysaccharide vaccines. This mutation occurs in the sixth exon of TWEAK and results in the amino acid substitution R145C within the conserved TNF-homology domain of the full-length protein. TWEAK mutant protein formed high molecular weight aggregates under nonreducing conditions, suggesting an increased propensity for intermolecular interactions. As a result, mutant TWEAK associated with B-cell–activating factor (BAFF) protein and down-regulated the BAFF-mediated activation of the noncanonical NF-κB pathway through inhibition of p100 processing to p52, resulting in inhibition of BAFF-dependent B-cell survival and proliferation. As BAFF mediates T-cell–independent isotype switching and B-cell survival, our data implicate TWEAK as a disease-susceptibility gene for a humoral immunodeficiency.


Nature Genetics | 2017

Germline hypomorphic CARD11 mutations in severe atopic disease

Chi A. Ma; Jeffrey R. Stinson; Yuan Zhang; Jordan K. Abbott; Michael Weinreich; Pia J Hauk; Paul R. Reynolds; Jonathan J. Lyons; Celeste G. Nelson; Elisa Ruffo; Batsukh Dorjbal; Salomé Glauzy; Natsuko Yamakawa; Swadhinya Arjunaraja; Kelsey Voss; Jennifer Stoddard; Julie E. Niemela; Yu Zhang; Sergio D. Rosenzweig; Joshua McElwee; Thomas DiMaggio; Helen F. Matthews; Nina Jones; Kelly D. Stone; Alejandro Palma; Matías Oleastro; Emma Prieto; Andrea Bernasconi; Geronimo Dubra; Silvia Danielian

Few monogenic causes for severe manifestations of common allergic diseases have been identified. Through next-generation sequencing on a cohort of patients with severe atopic dermatitis with and without comorbid infections, we found eight individuals, from four families, with novel heterozygous mutations in CARD11, which encodes a scaffolding protein involved in lymphocyte receptor signaling. Disease improved over time in most patients. Transfection of mutant CARD11 expression constructs into T cell lines demonstrated both loss-of-function and dominant-interfering activity upon antigen receptor–induced activation of nuclear factor-κB and mammalian target of rapamycin complex 1 (mTORC1). Patient T cells had similar defects, as well as low production of the cytokine interferon-γ (IFN-γ). The mTORC1 and IFN-γ production defects were partially rescued by supplementation with glutamine, which requires CARD11 for import into T cells. Our findings indicate that a single hypomorphic mutation in CARD11 can cause potentially correctable cellular defects that lead to atopic dermatitis.


Clinical Immunology | 2012

CD40 agonist antibody mediated improvement of chronic Cryptosporidium infection in patients with X-linked hyper IgM syndrome.

Xiying Fan; Bhaskar Upadhyaya; Liming Wu; Christopher Koh; Mónica Santín-Durán; Stefania Pittaluga; Gulbu Uzel; David E. Kleiner; Ester Williams; Chi A. Ma; Aaron Bodansky; Joao Bosco Oliveira; Pamela Edmonds; Ronald L. Hornung; Duane W. Wong; Ronald Fayer; Tom Fleisher; Theo Heller; Calman Prussin; Ashish Jain

X-linked hyper-IgM syndrome (XHM) is a combined immune deficiency disorder caused by mutations in CD40 ligand. We tested CP-870,893, a human CD40 agonist monoclonal antibody, in the treatment of two XHM patients with biliary Cryptosporidiosis. CP-870,893 activated B cells and APCs in vitro, restoring class switch recombination in XHM B cells and inducing cytokine secretion by monocytes. CP-870,893 infusions were well tolerated and showed significant activity in vivo, decreasing leukocyte concentration in peripheral blood. Although specific antibody responses were lacking, frequent dosing in one subject primed T cells to secrete IFN-g and suppressed oocyst shedding in the stool. Nevertheless, relapse occurred after discontinuation of therapy. The CD40 receptor was rapidly internalized following binding with CP-870,893, potentially explaining the limited capacity of CP-870,893 to mediate immune reconstitution. This study demonstrates that CP-870,893 suppressed oocysts shedding in XHM patients with biliary cryptosporidiosis. The continued study of CD40 agonists in XHM is warranted.


Journal of Experimental Medicine | 2017

ERBIN deficiency links STAT3 and TGF-β pathway defects with atopy in humans

Jonathan J. Lyons; Yihui Liu; Chi A. Ma; Xiaomin Yu; Michael P. O’Connell; Monica G. Lawrence; Yongqing Zhang; K. Karpe; Ming Zhao; A.M. Siegel; Kelly D. Stone; Celeste Nelson; N. Jones; Thomas DiMaggio; D.N. Darnell; E. Mendoza-Caamal; L. Orozco; Jason D. Hughes; Joshua McElwee; R.J. Hohman; P.A. Frischmeyer-Guerrerio; Marc E. Rothenberg; Alexandra F. Freeman; Steven M. Holland; Joshua D. Milner

Nonimmunological connective tissue phenotypes in humans are common among some congenital and acquired allergic diseases. Several of these congenital disorders have been associated with either increased TGF-&bgr; activity or impaired STAT3 activation, suggesting that these pathways might intersect and that their disruption may contribute to atopy. In this study, we show that STAT3 negatively regulates TGF-&bgr; signaling via ERBB2-interacting protein (ERBIN), a SMAD anchor for receptor activation and SMAD2/3 binding protein. Individuals with dominant-negative STAT3 mutations (STAT3mut) or a loss-of-function mutation in ERBB2IP (ERBB2IPmut) have evidence of deregulated TGF-&bgr; signaling with increased regulatory T cells and total FOXP3 expression. These naturally occurring mutations, recapitulated in vitro, impair STAT3–ERBIN–SMAD2/3 complex formation and fail to constrain nuclear pSMAD2/3 in response to TGF-&bgr;. In turn, cell-intrinsic deregulation of TGF-&bgr; signaling is associated with increased functional IL-4R&agr; expression on naive lymphocytes and can induce expression and activation of the IL-4/IL-4R&agr;/GATA3 axis in vitro. These findings link increased TGF-&bgr; pathway activation in ERBB2IPmut and STAT3mut patient lymphocytes with increased T helper type 2 cytokine expression and elevated IgE.

Collaboration


Dive into the Chi A. Ma's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jonathan J. Lyons

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Joshua D. Milner

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

Yongge Zhao

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kelly D. Stone

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Liming Wu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Steven M. Holland

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge