Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
J Pediatr Hematol Oncol ; 46(1): 39-45, 2024 01 01.
Article in English | MEDLINE | ID: mdl-38096154

ABSTRACT

Acute lymphoblastic leukemia treatment includes an outpatient (OP)-based 2-year maintenance therapy (MT). Over an 8-year period, patients were transited from only OP to a hybrid e-clinic/OP-clinic model. Electronic and patient-held medical records of acute lymphoblastic leukemia patients 1 to 18 years old during MT were used to analyze demographics, drug doses, treatment response and cost. A survey evaluated family satisfaction with the hybrid service. Four hundred and seventy-eight children, all with at least 1 year of MT from March 13, 2014 to March 24, 2022 were grouped into 4 treatment eras, representing the transition from all OP (era 1) to the current hybrid MT practice (era 4). Cohort demographics were similar across all eras. With transition to era 4, OP visits decreased to a third (16 to 18/48 visits). Practice optimization in era 2 resulted in higher MT dose intensity in subsequent eras (era 1: median 82% [interquartile range, 63 to 97]; era 2: 93% [73 to 108]; era 3: 88% [68 to 106]; era 4: 90% [74 to 114], P <0·0001), with no differences in absolute neutrophil count or neutropenia-related toxicity ( P =0.8). The hybrid service reduced MT expenses by ~50% and families (133/156, 85%) reported being very satisfied. Our experience indicates that a hybrid model is feasible, effective and less burdensome for patients and families.


Subject(s)
Electronic Mail , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Infant , Child, Preschool , Adolescent , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Leukocyte Count , Neutrophils
2.
J Allergy Clin Immunol ; 147(6): 2316-2329, 2021 06.
Article in English | MEDLINE | ID: mdl-33160971

ABSTRACT

BACKGROUND: The selective reduction of memory TH2 cell responses could be key to affording tolerance and protection from the recurrence of damaging allergic pathology. OBJECTIVE: We asked whether TNF family costimulatory molecules cooperated to promote accumulation and reactivity of effector memory CD4 T cells to inhaled complex allergen, and whether their neutralization could promote airway tolerance to subsequent reexposure to allergen. METHODS: Mice were sensitized intraperitoneally or intranasally with house dust mite and challenged with intranasal allergen after memory had developed. We assessed whether single or combined blockade of OX40L/CD252 and CD30L/CD153 inhibited memory T cells from driving acute asthmatic lung inflammation and protected mice following exposure to allergen at a later time. RESULTS: OX40- or CD30-deficient animals showed strong or partial protection against allergic airway inflammation; however, neutralizing either molecule alone during the secondary response to allergen had little effect on the frequency of effector memory CD4 T cells formed and acute lung inflammation. In contrast, a significant reduction in eosinophilic inflammation was observed when OX40L and CD30L were simultaneously neutralized, with dual blockade inhibiting effector memory TH2 cell expansion in the lungs, whereas formation of peripherally induced regulatory T cells remained intact. Moreover, dual blockade during the secondary response resulted in a tolerogenic state such that mice did not develop a normal tertiary memory TH2 cell and lung inflammatory response when challenged weeks later with allergen. CONCLUSION: Memory T-cell responses to complex allergens are controlled by several TNF costimulatory interactions, and their combination targeting might represent a strategy to reduce the severity of inflammatory reactions following reexposure to allergen.


Subject(s)
Allergens/immunology , CD30 Ligand/antagonists & inhibitors , Immunologic Memory , OX40 Ligand/antagonists & inhibitors , Th2 Cells/immunology , Th2 Cells/metabolism , Animals , Asthma/etiology , Asthma/metabolism , Asthma/pathology , Biomarkers , Disease Models, Animal , Disease Susceptibility/immunology , Humans , Hypersensitivity/immunology , Hypersensitivity/metabolism , Hypersensitivity/pathology , Mice , Pneumonia/etiology , Pneumonia/metabolism , Pneumonia/pathology
3.
Immunohorizons ; 4(8): 498-507, 2020 08 13.
Article in English | MEDLINE | ID: mdl-32792363

ABSTRACT

Respiratory virus infection normally drives neutrophil-dominated airway inflammation, yet some viral infections result in an eosinophil-dominated response in individuals such as allergic asthmatics. One idea is that viral infection simply exacerbates an ongoing type 2 response to allergen. However, prior exposure to allergen might alter the virus-induced innate response such that type 2-like eosinophilic inflammation can be induced. To test this, mice were sensitized intranasally with house dust mite allergen and then at later times exposed to rhinovirus RV1B via the airways. RV1B infection of naive mice led to the expected neutrophilic lung inflammatory response with no eosinophils or mucus production. In contrast, if mice were exposed to RV1B 1-4 wk after house dust mite inhalation, when the allergen response had subsided, infection led to eosinophilia and mucus production and a much stronger lymphocyte response that were partially or fully steroid resistant. In accordance, RV1B infection resulted in elevated expression of several inflammatory factors in allergen-pre-exposed mice, specifically those associated with type 2 immunity, namely CCL17, CXCL1, CCL2, IL-33, and IL-13. In vitro studies further showed that RV infection led to greater production of chemokines and cytokines in human bronchial epithelial cells that were previously stimulated with allergen, reinforcing the notion of an altered virus response after allergen exposure. In conclusion, we report that prior allergen exposure can modify responsiveness of cells in the lungs such that a qualitatively and quantitatively different inflammatory activity results following virus infection that is biased toward type 2-like airway disease.


Subject(s)
Eosinophils/metabolism , Lung Diseases/immunology , Picornaviridae Infections/immunology , Pyroglyphidae/immunology , Rhinovirus/physiology , Allergens/immunology , Animals , Chemokines/metabolism , Cytokines/metabolism , Inflammation/metabolism , Leukocyte Count , Lung Diseases/metabolism , Mice , Mice, Inbred C57BL , Rhinovirus/immunology
4.
Front Immunol ; 9: 576, 2018.
Article in English | MEDLINE | ID: mdl-29616048

ABSTRACT

The cytokine TNFSF14 [homologous to Lymphotoxin, exhibits Inducible expression and competes with HSV Glycoprotein D for binding to HVEM, a receptor expressed on T lymphocytes (LIGHT)] has been shown in mouse models to be important for development of lung tissue remodeling that is characteristic of asthma, idiopathic pulmonary fibrosis (IPF), and systemic sclerosis (SSc). However, its cellular targets are not fully delineated. In the present report, we show that LTßR and HVEM, the receptors for LIGHT, are constitutively expressed in primary human lung fibroblasts (HLFs). We asked whether LIGHT could promote inflammatory and remodeling-relevant activity in HLFs and how this was similar to, or distinct from, IL-13 or TGF-ß, two cytokines strongly implicated in the pathogenesis of asthma, IPF, and SSc. Accumulation of myofibroblasts expressing alpha smooth muscle actin is a feature of lung inflammatory diseases. LIGHT promoted cell cycle progression and proliferation of HLFs, but not alpha smooth muscle actin expression. In contrast, TGF-ß upregulated alpha smooth muscle actin but did not drive their proliferation. LIGHT also increased the gene or protein expression of a number of proinflammatory mediators, including ICAM-1 and VCAM-1, IL-6 and GM-CSF, the chemokines CCL5 and 20, and CXCL5, 11, and 12, and lung remodeling-associated proteinases MMP-9 and ADAM8. These were dependent on LTßR but not HVEM. LIGHT displayed overlapping and synergistic activities with IL-13 for a number of the activities, but LIGHT additionally enhanced the gene expression of several molecules, including the innate cytokines IL-33 and TSLP, which were not upregulated by IL-13. Our results highlight the varied and pleiotropic effects of LIGHT in HLFs. LIGHT might then be a therapeutic target for modulation of inflammation and remodeling associated with asthma and other similar diseases of the lung that involve fibroblasts.


Subject(s)
Fibroblasts/metabolism , Inflammation Mediators/metabolism , Interleukin-13/metabolism , Lung/metabolism , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Biomarkers , Cell Line , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/metabolism , Fibroblasts/drug effects , Gene Expression , Humans , Inflammation Mediators/pharmacology , Interleukin-13/genetics , Lung/pathology , Transforming Growth Factor beta/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/pharmacology
5.
Cytokine ; 101: 14-18, 2018 01.
Article in English | MEDLINE | ID: mdl-27531077

ABSTRACT

TNF (tumor necrosis factor) is both a pro-inflammatory and anti-inflammatory cytokine that is central to the development of autoimmune disease, cancer, and protection against infectious pathogens. As well as a myriad other activities, TNF can be a product of T cells and can act on T cells. Here we review old and new data on the importance of TNF produced by T cells and how TNF signaling via TNFR2 may directly impact alternate aspects of T cell biology. TNF can promote the activation and proliferation of naïve and effector T cells, but also can induce apoptosis of highly activated effector T cells, further determining the size of the pathogenic or protective conventional T cell pool. Moreover, TNF can have divergent effects on regulatory T cells. It can both downregulate their suppressive capacity, but also contribute in other instances to their development or accumulation. Biologics that block TNF or stimulate TNFR2 therefore have the potential to strongly modulate the balance between effector T cells and Treg cells which could impact disease in both positive and negative manners.


Subject(s)
T-Lymphocytes/immunology , Tumor Necrosis Factor-alpha/immunology , Animals , Apoptosis , Humans , Immunologic Memory , Inflammation , Mice , Receptors, Tumor Necrosis Factor, Type I/immunology , Receptors, Tumor Necrosis Factor, Type II/immunology , Signal Transduction/immunology , T-Lymphocytes/classification , T-Lymphocytes/physiology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor-alpha/biosynthesis
6.
J Immunol ; 199(8): 2721-2728, 2017 10 15.
Article in English | MEDLINE | ID: mdl-28877989

ABSTRACT

Stimulation of several TNF receptor family proteins has been shown to dampen inflammatory disease in murine models through augmenting the number and/or activity of regulatory T cells (Tregs). We recently found that one molecule, 4-1BB, used binding to Galectin-9 to exert its immunosuppressive effects and drive expansion of CD8+Foxp3- Tregs. We now show that ligation of another TNFR family molecule, DR3, which has previously been found to strongly expand CD4+Foxp3+ Tregs and suppress inflammation, also requires Galectin-9. We found that the extracellular region of DR3 directly binds to Galectin-9, and that Galectin-9 associates with DR3 in Tregs. From studies in vitro with Galectin-9-/- CD4+ T cells and Tregs, we found that stimulatory activity induced by ligating DR3 was in part dependent on Galectin-9. In vivo, in a model of experimental autoimmune encephalomyelitis, we show that an agonist of DR3 suppressed disease, correlating with expansion of CD4+Foxp3+ Tregs, and this protective effect was lost in Galectin-9-/- mice. Similar results were seen in an allergic lung inflammation model. Thus, we demonstrate a novel function of Galectin-9 in facilitating activity of DR3 related to Treg-mediated suppression.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Galectins/metabolism , Inflammation/immunology , Multiple Sclerosis/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism , Animals , Cell Proliferation , Cells, Cultured , Forkhead Transcription Factors/metabolism , Galectins/genetics , Humans , Immune Tolerance , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Receptors, Tumor Necrosis Factor, Member 25/metabolism , Signal Transduction
7.
J Allergy Clin Immunol ; 137(1): 278-288.e6, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26100084

ABSTRACT

BACKGROUND: Rhinovirus infection at an early age has been associated with development of asthma, but how rhinovirus influences the immune response is not clear. OBJECTIVE: Tolerance to inhaled antigen is mediated through induction of regulatory T (Treg) cells, and we examined whether rhinovirus infection of the respiratory tract can block airway tolerance by modulating Treg cells. METHODS: The immune response to intranasal ovalbumin in mice was assessed with concomitant infection with RV1B, and the factors induced in vivo were compared with those made by human lung epithelial cells infected in vitro with RV16. RESULTS: RV1B infection of mice abrogated tolerance induced by inhalation of soluble ovalbumin, suppressing the normal generation of forkhead box protein 3-positive Treg cells while promoting TH2 cells. Furthermore, RV1B infection led to susceptibility to asthmatic lung disease when mice subsequently re-encountered aeroantigen. RV1B promoted early in vivo expression of the TNF family protein OX40 ligand on lung dendritic cells that was dependent on the innate cytokine thymic stromal lymphopoietin (TSLP) and also induced another innate cytokine, IL-33. Inhibiting each of these pathways allowed the natural development of Treg cells while minimizing TH2 differentiation and restored tolerance in the face of RV1B infection. In accordance, RV16 infection of human lung epithelial cells upregulated TSLP and IL-33 expression. CONCLUSIONS: These results suggest that infection of the respiratory epithelium with rhinovirus can antagonize tolerance to inhaled antigen through combined induction of TSLP, IL-33, and OX40 ligand and that this can lead to susceptibility to asthmatic lung inflammation.


Subject(s)
Cytokines/immunology , Immune Tolerance , Interleukin-33/immunology , Membrane Glycoproteins/immunology , Picornaviridae Infections/immunology , Rhinovirus , Tumor Necrosis Factors/immunology , Animals , Antigens/immunology , Bronchoalveolar Lavage Fluid/immunology , Epithelial Cells , Humans , Interleukin-13/immunology , Interleukin-4/immunology , Lung/cytology , Mice, Inbred C57BL , Mice, Transgenic , OX40 Ligand , Ovalbumin/immunology , Respiratory Hypersensitivity/immunology , T-Lymphocytes/immunology , Thymic Stromal Lymphopoietin
8.
J Immunol ; 187(12): 6499-507, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22079987

ABSTRACT

Regulatory T cells (Tregs) play a critical role in the maintenance of airway tolerance. We report that inhaled soluble Ag induces adaptive Foxp3(+) Tregs, as well as a regulatory population of CD4(+) T cells in the lungs and lung-draining lymph nodes that express latency-associated peptide (LAP) on their cell surface but do not express Foxp3. Blocking the cytokine IL-10 or TGF-ß prevented the generation of LAP(+) Tregs and Foxp3(+) Tregs in vivo, and the LAP(+) Tregs could also be generated concomitantly with Foxp3(+) Tregs in vitro by culturing naive CD4(+) T cells with Ag and exogenous TGF-ß. The LAP(+) Tregs strongly suppressed naive CD4(+) T cell proliferation, and transfer of sorted OVA-specific LAP(+) Tregs in vivo inhibited allergic eosinophilia and Th2 cytokine expression in the lung, either when present at the time of Th2 sensitization or when injected after Th2 cells were formed. Furthermore, inflammatory innate stimuli from house dust mite extract, nucleotide-binding oligomerization domain containing 2 ligand, and LPS, which are sufficient for blocking airway tolerance, strongly decreased the induction of LAP(+) Tregs. Taken together, we concluded that inducible Ag-specific LAP(+) Tregs can suppress asthmatic lung inflammation and constitute a mediator of airway tolerance together with Foxp3(+) Tregs.


Subject(s)
Cell Differentiation/immunology , Forkhead Transcription Factors/deficiency , Respiratory Hypersensitivity/pathology , Respiratory Hypersensitivity/prevention & control , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/biosynthesis , Allergens/administration & dosage , Allergens/physiology , Animals , Cell Differentiation/genetics , Cells, Cultured , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/genetics , Forkhead Transcription Factors/genetics , Genes, Reporter , Immune Tolerance/genetics , Inflammation/immunology , Inflammation/pathology , Inflammation/prevention & control , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/administration & dosage , Ovalbumin/physiology , Respiratory Hypersensitivity/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta1/genetics
9.
J Allergy Clin Immunol ; 126(6): 1284-93.e10, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21051079

ABSTRACT

BACKGROUND: Airway tolerance is critical for protecting the lung from inflammatory disease driven by allergens. However, factors that disrupt tolerance processes and then lead to susceptibility to developing allergic asthma remain elusive. OBJECTIVE: To investigate whether recognition of bacterial microbial-associated molecular patterns in the lung may result in susceptibility to developing allergic reactions, and to understand the molecular mechanisms by which such triggers block natural tolerance. METHODS: Ligands of intracellular microbial-associated molecular pattern recognition receptors-the nucleotide-binding oligomerization domain (Nod)-like receptors, Nod1 and Nod2-were given intranasally with antigen, and their ability to modulate airway tolerance was analyzed. RESULTS: Intranasal Nod2 ligand rapidly induced lung expression of the innate cytokines thymic stromal lymphopoietin and IL-25, and thymic stromal lymphopoietin promoted expression of OX40 ligand, a T-cell-costimulatory ligand, on lung CD11c(+)CD11b(+) cells and B220(+) cells. Together these 3 molecules blocked the generation of antigen-specific CD4(+)forkhead box protein 3(+) adaptive regulatory T cells and concomitantly drove IL-4-producing CD4 T cells. By altering the regulatory T/T(H)2-cell balance, tolerance was blocked, and sensing of Nod2 ligand resulted in subsequent susceptibility to developing eosinophil-dominated airway inflammation. CONCLUSION: We show that a Nod-like receptor is a novel, previously unrecognized, pathway that adversely links innate and adaptive immunity and leads to allergic disease and asthmatic lung inflammation.


Subject(s)
Hypersensitivity/immunology , Lung/metabolism , Nod2 Signaling Adaptor Protein/administration & dosage , T-Lymphocytes, Regulatory/metabolism , Th2 Cells/metabolism , Acetylmuramyl-Alanyl-Isoglutamine/administration & dosage , Acetylmuramyl-Alanyl-Isoglutamine/pharmacology , Allergens/administration & dosage , Animals , Cells, Cultured , Cytokines/biosynthesis , Cytokines/genetics , Eosinophilia , Hypersensitivity/drug therapy , Hypersensitivity/pathology , Hypersensitivity/physiopathology , Immune Tolerance/drug effects , Immunity, Innate/immunology , Interleukin-17/biosynthesis , Interleukin-17/genetics , Lung/immunology , Lung/pathology , Mice , Mice, Inbred C57BL , Nod2 Signaling Adaptor Protein/agonists , OX40 Ligand/biosynthesis , OX40 Ligand/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th2 Cells/drug effects , Th2 Cells/immunology , Th2 Cells/pathology , Thymic Stromal Lymphopoietin
10.
Immunobiology ; 215(7): 527-34, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19897276

ABSTRACT

Asthma is a chronic immune inflammatory disease characterized by variable airflow obstruction and increased bronchial hyperreactivity (BHR). Therapeutic interventions reduce airway inflammation and relieve symptoms but associated with potential side effects that limit their usefulness. The present study was undertaken to assess the effect of choline on immune inflammation and BHR in asthma subjects. The patients of asthma (n=76) were recruited and treated with choline supplement (1500 mg twice) or standard pharmacotherapy for 6 months in two groups. The patients were evaluated by clinical, immunologic and biochemical parameters. The treatment with choline showed significant reduction in symptom/drug score and improvement in PC(20) FEV1 compared to baseline or standard pharmacotherapy (p<0.01). Choline therapy significantly reduced IL-4, IL-5 and TNF-alpha level as compared to baseline or standard pharmacotherapy after 6 months (p<0.01). Blood eosinophil count and total IgE levels were reduced in both the treatment groups. Cysteinyl leukotriene and leukotriene B4 were suppressed significantly by choline treatment (p<0.01). This was accompanied by decreased 8-isoprostanes, a biomarker for oxidative stress after choline treatment (p<0.01). Choline therapy modulates immune inflammation and suppresses oxidative stress in asthma patients. It can be used as an adjunct therapy for asthma patients.


Subject(s)
Asthma/immunology , Choline/pharmacology , Eosinophils/drug effects , Adolescent , Adult , Asthma/diagnosis , Asthma/physiopathology , Bronchial Hyperreactivity , Cells, Cultured , Cytokines/metabolism , Eosinophils/pathology , Female , Follow-Up Studies , Forced Expiratory Volume , Humans , Immunosuppression Therapy , Inflammation , Leukotrienes/metabolism , Male , Middle Aged , Oxidative Stress/drug effects , Oxidative Stress/immunology , Skin Tests , Th2 Cells/drug effects , Th2 Cells/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...