Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters











Publication year range
1.
Mucosal Immunol ; 12(5): 1201-1211, 2019 09.
Article in English | MEDLINE | ID: mdl-31417161

ABSTRACT

Uncontrolled interferon γ (IFNγ)-mediated T-cell responses to commensal microbiota are a driver of inflammatory bowel disease (IBD). Interleukin-10 (IL-10) is crucial for controlling these T-cell responses, but the precise mechanism of inhibition remains unclear. A better understanding of how IL-10 exerts its suppressive function may allow identification of individuals with suboptimal IL-10 function among the heterogeneous population of IBD patients. Using cells from patients with an IL10RA deficiency or STAT3 mutations, we demonstrate that IL-10 signaling in monocyte-derived dendritic cells (moDCs), but not T cells, is essential for controlling IFNγ-secreting CD4+ T cells. Deficiency in IL-10 signaling dramatically increased IL-1ß release by moDCs. IL-1ß boosted IFNγ secretion by CD4+ T cells either directly or indirectly by stimulating moDCs to secrete IL-12. As predicted a signature of IL-10 dysfunction was observed in a subgroup of pediatric IBD patients having higher IL-1ß expression in activated immune cells and macroscopically affected intestinal tissue. In agreement, reduced IL10RA expression was detected in peripheral blood mononuclear cells and a subgroup of pediatric IBD patients exhibited diminished IL-10 responsiveness. Our data unveil an important mechanism by which IL-10 controls IFNγ-secreting CD4+ T cells in humans and identifies IL-1ß as a potential classifier for a subgroup of IBD patients.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Interferon-gamma/metabolism , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Signal Transduction , Adolescent , Cell Communication , Child , Disease Susceptibility , Humans , Inflammatory Bowel Diseases/etiology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/therapy
3.
J Exp Med ; 214(3): 669-680, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28126831

ABSTRACT

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-ß 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-ß 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-ß 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-ß. In turn, cell-intrinsic deregulation of TGF-ß signaling is associated with increased functional IL-4Rα expression on naive lymphocytes and can induce expression and activation of the IL-4/IL-4Rα/GATA3 axis in vitro. These findings link increased TGF-ß pathway activation in ERBB2IPmut and STAT3mut patient lymphocytes with increased T helper type 2 cytokine expression and elevated IgE.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Hypersensitivity/immunology , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Adaptor Proteins, Signal Transducing/deficiency , Humans , Interleukin-4/physiology , Receptors, Interleukin-4/physiology , Smad2 Protein/analysis , Smad2 Protein/physiology , Smad3 Protein/analysis , Smad3 Protein/physiology
4.
J Clin Pharm Ther ; 42(1): 75-79, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27982447

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: Posaconazole is an extended-spectrum triazole antifungal with activity against a variety of clinically significant yeasts and moulds. Posaconazole is not currently approved by the U.S. Food and Drug Administration for use in children younger than 13 years of age. Our primary objective was to describe the dosing and observed trough concentrations with posaconazole oral suspension in paediatric patients at the National Institutes of Health Clinical Center (Bethesda, MD). METHODS: This retrospective single-centre study reviewed paediatric patients younger than 13 years of age initiated on posaconazole oral suspension. Patients were included if they were initiated on posaconazole for prophylaxis or treatment for fungal infections from September 2006 through March 2013 with at least one trough concentration collected after at least 7 days of therapy. RESULTS AND DISCUSSION: A total of 20 male patients were included, of whom 15 (75%) had chronic granulomatous disease. The median age of patients was 6·5 years (range: 2·8-10·7). A total of 79 posaconazole trough concentrations were measured in patients receiving posaconazole as prophylaxis (n = 8) or treatment (n = 12). Posaconazole dose referenced to total body weight ranged from 10·0 to 49·2 mg/kg/day. Posaconazole trough concentrations ranged from undetectable (<50 ng/mL) up to 3620 ng/mL and were ≥500, ≥700 and ≥1250 ng/mL in 95%, 60% and 25% of patients, respectively. WHAT IS NEW AND CONCLUSIONS: Patients younger than 13 years of age had highly variable trough concentrations, and recommendations for the appropriate dosing of posaconazole oral suspension remain challenging. Until studies are conducted to determine the appropriate dosing of posaconazole in this patient population, therapeutic drug monitoring should be considered to ensure adequate posaconazole exposure.


Subject(s)
Antifungal Agents/administration & dosage , Mycoses/drug therapy , Suspensions/administration & dosage , Triazoles/administration & dosage , Administration, Oral , Child , Child, Preschool , Drug Monitoring/methods , Humans , Male , Retrospective Studies
5.
J Clin Pharm Ther ; 40(5): 609-611, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26248976

ABSTRACT

WHAT IS KNOWN AND OBJECTIVE: Despite the known significant drug-drug interaction between isavuconazole and tacrolimus, there are no recommendations on dose adjustment when these drugs are given concomitantly. We report on a patient with a mediastinal Aspergillus fumigatus infection resistant to posaconazole and describe how she was successfully managed with tacrolimus therapeutic drug-level monitoring. CASE SUMMARY: Our patient presented with a mediastial Aspergillus fumigatus infection, 2 years after lung transplantation. A. fumigatus was resistant to posaconazole, and the patient had intolerance to voriconazole shown by elevated transaminases. The patient was given isavuconazole with drug-level monitoring. She was managed successfully with no adverse events. Tacrolimus concentration continued to increase after more than 2 weeks of therapy and required a further reduction to 72% of the usual dose to maintain the target concentrations over a 8-week period. WHAT IS NEW AND CONCLUSION: When isavuconazole is given to patients on tacrolimus, the dose of the latter will need considerable reduction. We would suggest an initial 50% reduction and recommend close weekly monitoring of tacrolimus concentration. Further dose decreases of 25-50% may be required.

6.
J Clin Virol ; 63: 46-50, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25600604

ABSTRACT

We present a 17-year old girl with DOCK-8 deficiency, severe untreated oral HSV-1 infection and associated aggressive periodontitis. DOCK-8 deficiency is a primary immunodeficiency, caused by biallelicloss-of-function mutations in the DOCK8 gene, often leading to severe viral and fungal mucocutaneous infections. Nevertheless, to date DOCK8 has not been associated with severe periodontitis and inflammatory bone loss around teeth. Understanding whether DOCK8 deficiency or severe HSV-1 infection underlies susceptibility to periodontitis is central to this case and may provide insights into susceptibility factors for periodontitis in the general population. Our clinical and microbiological data suggest that severe HSV-1 infection is the driver of periodontal inflammation in this case.


Subject(s)
Aggressive Periodontitis/pathology , Aggressive Periodontitis/virology , Guanine Nucleotide Exchange Factors/deficiency , Herpes Simplex/complications , Herpes Simplex/diagnosis , Herpesvirus 1, Human/isolation & purification , Adolescent , Disease Susceptibility , Female , Herpes Simplex/pathology , Humans
7.
J Clin Immunol ; 33(5): 991-1001, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23512243

ABSTRACT

Patients with deficiency in the interferon gamma receptor (IFN-γR) are unable to respond properly to IFN-γ and develop severe infections with nontuberculous mycobacteria (NTM). IFN-γ and IFN-α are known to signal through STAT1 and activate many downstream effector genes in common. Therefore, we added IFN-α for treatment of patients with disseminated mycobacterial disease in an effort to complement their IFN-γ signaling defect. We treated four patients with IFN-γR deficiency with adjunctive IFN-α therapy in addition to best available antimicrobial therapy, with or without IFN-γ, depending on the defect. During IFN-α treatment, ex vivo induction of IFN target genes was detected. In addition, IFN-α driven gene expression in patients' cells and mycobacteria induced cytokine response were observed in vitro. Clinical responses varied in these patients. IFN-α therapy was associated with either improvement or stabilization of disease. In no case was disease exacerbated. In patients with profoundly impaired IFN-γ signaling who have refractory infections, IFN-α may have adjunctive anti-mycobacterial effects.


Subject(s)
Interferon-alpha/therapeutic use , Interferon-gamma/metabolism , Receptors, Interferon/metabolism , Adult , Cells, Cultured , Child, Preschool , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression/drug effects , Humans , Interferon-gamma/genetics , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Mycobacterium/genetics , Mycobacterium/metabolism , Mycobacterium Infections/genetics , Mycobacterium Infections/metabolism , Receptors, Interferon/genetics , Signal Transduction/drug effects , Young Adult , Interferon gamma Receptor
8.
Mucosal Immunol ; 4(4): 448-55, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21346738

ABSTRACT

Oropharyngeal candidiasis (OPC, thrush) is an opportunistic infection caused by the commensal fungus Candida albicans. An understanding of immunity to Candida has recently begun to unfold with the identification of fungal pattern-recognition receptors such as C-type lectin receptors, which trigger protective T-helper (Th)17 responses in the mucosa. Hyper-IgE syndrome (HIES/Job's syndrome) is a rare congenital immunodeficiency characterized by dominant-negative mutations in signal transducer and activator of transcription 3, which is downstream of the Th17-inductive cytokines interleukin (IL)-6 and IL-23, and hence patients with HIES exhibit dramatic Th17 deficits. HIES patients develop oral and mucocutaneous candidiasis, supporting a protective role for Th17 cells in immunity to OPC. However, the Th17-dependent mechanisms of antifungal immunity in OPC are still poorly defined. An often unappreciated aspect of oral immunity is saliva, which is rich in antimicrobial proteins (AMPs) and exerts direct antifungal activity. In this study, we show that HIES patients show significant impairment in salivary AMPs, including ß-defensin 2 and Histatins. This tightly correlates with reduced candidacidal activity of saliva and concomitantly elevated colonization with Candida. Moreover, IL-17 induces histatins in cultured salivary gland cells. This is the first demonstration that HIES is associated with defective salivary activity, and provides a mechanism for the severe susceptibility of these patients to OPC.


Subject(s)
Candidiasis/complications , Candidiasis/immunology , Job Syndrome/complications , Job Syndrome/immunology , Mouth Mucosa/immunology , Mouth Mucosa/microbiology , Adenosine Monophosphate/immunology , Adenosine Monophosphate/metabolism , Adolescent , Adult , Candida albicans/immunology , Child , Female , Gene Expression Regulation/immunology , Histatins/immunology , Histatins/metabolism , Humans , Immunity, Mucosal , Male , Middle Aged , Saliva/immunology , Th17 Cells/immunology , Th17 Cells/metabolism , Young Adult , beta-Defensins/immunology , beta-Defensins/metabolism
9.
Oral Dis ; 15(1): 2-7, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19036057

ABSTRACT

Autosomal dominant hyper IgE (HIES or Job's) syndrome is a rare primary immune deficiency characterized by eczema, recurrent skin and lung infections, extremely elevated serum IgE, and a variety of connective tissue and skeletal abnormalities. Individuals with HIES share a characteristic facial appearance and many oral manifestations including retained primary dentition, a high arched palate, variations of the oral mucosa and gingiva, and recurrent oral candidiasis. Mutations in STAT3 account for the majority, if not all, of the cases of autosomal dominant HIES, but the pathogenesis of the many varied features remains poorly understood. In this review, we discuss the clinical phenotype of HIES including immunologic and non-immunologic features, the genetics of HIES, and treatment.


Subject(s)
Job Syndrome/immunology , Mouth Diseases/immunology , Tooth Diseases/immunology , Candidiasis, Oral/immunology , Facies , Gingival Diseases/immunology , Humans , Job Syndrome/genetics , Mouth Mucosa/pathology , Mutation/genetics , Palate/pathology , Phenotype , Recurrence , STAT3 Transcription Factor/genetics , Tooth, Deciduous/pathology
10.
Oral Dis ; 14(1): 73-81, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18173452

ABSTRACT

AIM: Hyperimmunoglobulin-E syndrome (HIES) is a primary immunodeficiency characterized by eczema, recurrent skin and lung infections with pneumatocoele formation, and extremely elevated serum immunoglobulin-E. The precise immunologic defect and genetic etiology remain unknown. Non-immunologic findings include characteristic facial features (prominent forehead, fleshy nasal tip, and increased interalar distance); skeletal involvement (pathological fractures, scoliosis, and craniosynostosis); and retention of primary teeth. This study aims to characterize intraoral soft tissue findings in HIES patients. METHODS: Sixty HIES patients (4-54 years, 27 males, 33 females) received intraoral and radiographic evaluations. Chronological dental development was also assessed. RESULTS: Lesions of the hard palate and dorsal tongue were found in 55% and 60% of patients, respectively. Palatal lesions ranged from a generalized surface keratosis to a midline sagittal fibrotic bridge. Tongue lesions consisted of multiple fissures and a midline cleft. On the lip and buccal mucosa, keratotic plaques and/or surface fissures were found in 8% and 23% of patients, respectively. Manifested in 76.7% of patients, the intraoral lesions were significantly more prevalent than the characteristic facial traits (P=0.0013). CONCLUSIONS: Alterations in oral mucosa and gingiva were present in the majority of HIES patients. These novel intraoral findings may facilitate the diagnosis of HIES.


Subject(s)
Hypergammaglobulinemia/immunology , Immunoglobulin E/immunology , Mouth Diseases/immunology , Adolescent , Adult , Child , Child, Preschool , Female , Fibrosis , Humans , Leukoplakia, Oral/immunology , Lip Diseases/immunology , Male , Middle Aged , Mouth Mucosa/immunology , Odontogenesis/immunology , Palate, Hard/immunology , Phenotype , Syndrome , Tongue/abnormalities , Tongue Diseases/immunology
11.
Pediatr Cardiol ; 26(5): 578-84, 2005.
Article in English | MEDLINE | ID: mdl-16132289

ABSTRACT

Angiogenesis has been shown to be dysregulated in coronary artery (CA) aneurysms in the chronic phase of Kawasaki disease (KD). Neovascularization may occur in inflammatory-related vascular diseases because many angiogenesis mediators are secreted by inflammatory cells. We hypothesized that inflammation of the acute KD CA aneurysm could lead to dysregulation of angiogenesis mediators and subsequent neovascularization. To investigate this hypothesis, acute fatal KD cardiac tissues were immunostained for angiogenic inducers and inhibitors. Microvessel density was determined and the degree of inflammation assessed. Marked inflammation and angiogenesis were found in acute KD CA aneurysms and myocardium, with the highest microvessel density seen in patients who died 2-3 weeks after onset of the disease. Expression of proangiogenic proteins was higher than expression of inhibitors in KD CA aneurysms and myocardium. Angiogenesis mediators were localized to inflammatory cells in the myointima, adventitia, and myocardium. We conclude that significant neovascularization occurs in acute KD CA aneurysms and myocardium much sooner after onset of the disease than has been previously reported, that multiple angiogenesis factors are involved, and that dysregulation of angiogenesis likely contributes to KD vasculopathy.


Subject(s)
Coronary Aneurysm/mortality , Coronary Vessels/pathology , Mucocutaneous Lymph Node Syndrome/mortality , Mucocutaneous Lymph Node Syndrome/pathology , Myocardium/pathology , Neovascularization, Pathologic/mortality , Acute Disease , Aneurysm, Ruptured/mortality , Angiostatins/metabolism , Case-Control Studies , Child , Coronary Aneurysm/metabolism , Coronary Aneurysm/pathology , Coronary Vessels/metabolism , Female , Fibroblast Growth Factor 2/metabolism , Humans , Immunohistochemistry , Infant , Infant, Newborn , Male , Mast Cells/metabolism , Microcirculation , Mucocutaneous Lymph Node Syndrome/metabolism , Myocarditis/metabolism , Myocarditis/mortality , Myocarditis/pathology , Myocardium/metabolism , Neovascularization, Pathologic/metabolism , Nerve Growth Factor/metabolism , Platelet-Derived Growth Factor/metabolism , Thrombospondins/metabolism , Vascular Endothelial Growth Factor A/metabolism
12.
Curr Opin Infect Dis ; 14(3): 357-61, 2001 Jun.
Article in English | MEDLINE | ID: mdl-11964855

ABSTRACT

Kawasaki disease is now being recognized as the leading cause of acquired heart disease in children in North America and Japan. This review discusses the recent developments and controversies in Kawasaki disease. Increasing evidence has supported an infectious etiology for Kawasaki disease; however, the debate continues as to whether the inflammatory response results from a conventional antigen or a superantigen. Recent immunohistochemistry findings suggest many vascular growth factors then play a role in the formation of the coronary artery lesions. Many studies have focused on the identification and therapy for patients resistant to conventional therapy, as well as the long-term prognosis of Kawasaki disease survivors. The recent advances in Kawasaki disease are helping to provide some clues in the etiology, pathogenesis and therapy for Kawasaki disease.


Subject(s)
Mucocutaneous Lymph Node Syndrome , Child , Child, Preschool , Coronary Aneurysm/physiopathology , Humans , Immunoglobulins, Intravenous/therapeutic use , Mucocutaneous Lymph Node Syndrome/etiology , Mucocutaneous Lymph Node Syndrome/physiopathology , Mucocutaneous Lymph Node Syndrome/therapy
SELECTION OF CITATIONS
SEARCH DETAIL