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











Publication year range
1.
Pediatr Blood Cancer ; : e31348, 2024 Sep 25.
Article in English | MEDLINE | ID: mdl-39323047

ABSTRACT

BACKGROUND: Hydroxyurea remains underutilized in the pediatric sickle cell population despite its well-known efficacy in decreasing sickle cell complications and hospitalizations. Access to refills and liquid formulation remains a critical barrier to adherence to hydroxyurea regimens. This study was undertaken to determine the clinical impact of home-delivering compounded liquid hydroxyurea (LHU) to pediatric patients with sickle cell disease. PROCEDURE/METHODS: A retrospective cohort study was conducted using electronic health records and pharmacy databases. Pediatric patients younger than 21 years of age at the time of hydroxyurea initiation from March 2016 to July 2020 who received compounded LHU from Boston Medical Center Pharmacy were included. The primary outcomes of the study were drug adherence (assessed by evaluating the proportion of days covered [PDC]), rates of acute care utilization before and after enrolling in the LHU delivery program. RESULTS: The final cohort included 41 patients, showing a significant decrease in hospitalizations (p = .01) and acute chest syndrome episodes (p = .03) after the initiation of the LHU delivery program. In comparing hydroxyurea-naïve patients with those previously exposed to hydroxyurea, the latter group had lower hospitalization rates (p = .01), fewer vaso-occlusive event (VOE) episodes (p = .02), and fewer emergency department (ED) visits (p = .01). The median PDC value 1 year post initiation of LHU was 95. CONCLUSIONS: Home delivery of compounded LHU from the pharmacy to pediatric sickle cell disease patients improved access to hydroxyurea, and was linked to reduced hospitalizations and acute chest syndrome episodes. This advancement in cost savings and improved patient outcomes is a significant step forward in pediatric hematology. By overcoming access barriers, home delivery programs can greatly enhance outcomes among pediatric patients with sickle cell disease.

3.
Ann Allergy Asthma Immunol ; 133(2): 144-149, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38750723

ABSTRACT

Medical ethics is relevant to the clinical practice of allergy and immunology regardless of the type of patient, disease state, or practice setting. When engaging in clinical care, performing research, or enacting policies on the accessibility and distribution of healthcare resources, physicians regularly make and justify decisions using the fundamental principles of medical ethics. Thus, knowledge of these principles is paramount for allergists/immunologists. To date, there has been a shortage of medical ethics research in allergy and immunology. This review describes this scarcity, highlights publication trends over time, and advocates for additional support for research and training in medical ethics with a focus on topics germane to the practice of allergy and immunology.


Subject(s)
Allergy and Immunology , Humans , Allergy and Immunology/ethics , Ethics, Medical , Biomedical Research/ethics
4.
Immunol Allergy Clin North Am ; 44(2): 281-291, 2024 May.
Article in English | MEDLINE | ID: mdl-38575223

ABSTRACT

Immunotherapy is a treatment approach based on the principle of incremental allergen exposure to achieve desensitization. Recently, oral immunotherapy has been introduced as a treatment of IgE-mediated food allergy. Some patients receiving oral immunotherapy for food allergy may develop eosinophilic esophagitis. Here, we summarize the literature examining this association, its treatment, and outcomes and discuss possible explanations for this clinical phenomenon. We further identify potential associations with aeroallergen sensitivity and other forms of immunotherapy including subcutaneous immunotherapy and sublingual immunotherapy. Finally, we discuss management of immunotherapy-induced eosinophilic esophagitis. Epicutaneous immunotherapy is highlighted as an area of therapeutic investigation.


Subject(s)
Eosinophilic Esophagitis , Food Hypersensitivity , Sublingual Immunotherapy , Humans , Eosinophilic Esophagitis/etiology , Eosinophilic Esophagitis/therapy , Desensitization, Immunologic/adverse effects , Food Hypersensitivity/drug therapy , Allergens/therapeutic use
6.
Nat Commun ; 11(1): 2286, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32385332

ABSTRACT

Studies on macrophage gene expression have historically focused on events leading to RNA polymerase II recruitment and transcription initiation, whereas the contribution of post-initiation steps to macrophage activation remains poorly understood. Here, we report that widespread promoter-proximal RNA polymerase II pausing in resting macrophages is marked by co-localization of the negative elongation factor (NELF) complex and facilitated by PU.1. Upon inflammatory stimulation, over 60% of activated transcriptome is regulated by polymerase pause-release and a transient genome-wide NELF dissociation from chromatin, unexpectedly, independent of CDK9, a presumed NELF kinase. Genetic disruption of NELF in macrophages enhanced transcription of AP-1-encoding Fos and Jun and, consequently, AP-1 targets including Il10. Augmented expression of IL-10, a critical anti-inflammatory cytokine, in turn, attenuated production of pro-inflammatory mediators and, ultimately, macrophage-mediated inflammation in vivo. Together, these findings establish a previously unappreciated role of NELF in constraining transcription of inflammation inhibitors thereby enabling inflammatory macrophage activation.


Subject(s)
Anti-Inflammatory Agents/metabolism , Gene Expression Regulation , Inflammation/genetics , Macrophages/pathology , Transcription Factors/metabolism , Animals , Chromatin/metabolism , Interleukin-10/metabolism , Macrophage Activation/genetics , Macrophages/metabolism , Mice , Nucleotide Motifs/genetics , Promoter Regions, Genetic , RNA Polymerase II/metabolism , Transcription Initiation Site , Transcription, Genetic , Transcriptional Activation/genetics
7.
Elife ; 72018 02 09.
Article in English | MEDLINE | ID: mdl-29424686

ABSTRACT

The glucocorticoid receptor (GR) potently represses macrophage-elicited inflammation, however, the underlying mechanisms remain obscure. Our genome-wide analysis in mouse macrophages reveals that pro-inflammatory paused genes, activated via global negative elongation factor (NELF) dissociation and RNA Polymerase (Pol)2 release from early elongation arrest, and non-paused genes, induced by de novo Pol2 recruitment, are equally susceptible to acute glucocorticoid repression. Moreover, in both cases the dominant mechanism involves rapid GR tethering to p65 at NF-kB-binding sites. Yet, specifically at paused genes, GR activation triggers widespread promoter accumulation of NELF, with myeloid cell-specific NELF deletion conferring glucocorticoid resistance. Conversely, at non-paused genes, GR attenuates the recruitment of p300 and histone acetylation, leading to a failure to assemble BRD4 and Mediator at promoters and enhancers, ultimately blocking Pol2 initiation. Thus, GR displays no preference for a specific pro-inflammatory gene class; however, it effects repression by targeting distinct temporal events and components of transcriptional machinery.


Subject(s)
Gene Expression Regulation , Inflammation , Macrophages/immunology , Receptors, Glucocorticoid/metabolism , Transcription, Genetic , Animals , Cells, Cultured , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic
8.
Nat Commun ; 8(1): 1739, 2017 11 23.
Article in English | MEDLINE | ID: mdl-29170386

ABSTRACT

The glucocorticoid (GC) receptor (GR) suppresses inflammation by activating anti-inflammatory and repressing pro-inflammatory genes. GR-interacting protein-1 (GRIP1) is a GR corepressor in macrophages, however, whether GRIP1 mediates GR-activated transcription, and what dictates its coactivator versus corepressor properties is unknown. Here we report that GRIP1 loss in macrophages attenuates glucocorticoid induction of several anti-inflammatory targets, and that GC treatment of quiescent macrophages globally directs GRIP1 toward GR binding sites dominated by palindromic GC response elements (GRE), suggesting a non-redundant GRIP1 function as a GR coactivator. Interestingly, GRIP1 is phosphorylated at an N-terminal serine cluster by cyclin-dependent kinase-9 (CDK9), which is recruited into GC-induced GR:GRIP1:CDK9 hetero-complexes, producing distinct GRE-specific GRIP1 phospho-isoforms. Phosphorylation potentiates GRIP1 coactivator but, remarkably, not its corepressor properties. Consistently, phospho-GRIP1 and CDK9 are not detected at GR transrepression sites near pro-inflammatory genes. Thus, GR restricts actions of its own coregulator via CDK9-mediated phosphorylation to a subset of anti-inflammatory genes.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Cyclin-Dependent Kinase 9/metabolism , Glucocorticoids/metabolism , Macrophages/metabolism , Nerve Tissue Proteins/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/genetics , Animals , Binding Sites/genetics , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/genetics , Cell Line , Cells, Cultured , Dexamethasone/pharmacology , Gene Knockdown Techniques , Glucocorticoids/pharmacology , Humans , Inflammation/genetics , Inflammation/metabolism , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Phosphorylation , Receptors, Glucocorticoid/metabolism , Response Elements , Transcriptional Activation
9.
Nat Commun ; 7: 12254, 2016 07 28.
Article in English | MEDLINE | ID: mdl-27464507

ABSTRACT

Diet-induced obesity causes chronic macrophage-driven inflammation in white adipose tissue (WAT) leading to insulin resistance. WAT macrophages, however, differ in their origin, gene expression and activities: unlike infiltrating monocyte-derived inflammatory macrophages, WAT-resident macrophages counteract inflammation and insulin resistance, yet, the mechanisms underlying their transcriptional programming remain poorly understood. We recently reported that a nuclear receptor cofactor-glucocorticoid receptor (GR)-interacting protein (GRIP)1-cooperates with GR to repress inflammatory genes. Here, we show that GRIP1 facilitates macrophage programming in response to IL4 via a GR-independent pathway by serving as a coactivator for Kruppel-like factor (KLF)4-a driver of tissue-resident macrophage differentiation. Moreover, obese mice conditionally lacking GRIP1 in macrophages develop massive macrophage infiltration and inflammation in metabolic tissues, fatty livers, hyperglycaemia and insulin resistance recapitulating metabolic disease. Thus, GRIP1 is a critical regulator of immunometabolism, which engages distinct transcriptional mechanisms to coordinate the balance between macrophage populations and ultimately promote metabolic homeostasis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Polarity , Homeostasis , Macrophages/cytology , Macrophages/metabolism , Nerve Tissue Proteins/metabolism , Trans-Activators/metabolism , Adaptor Proteins, Signal Transducing/deficiency , Animals , Cell Line , Cell Polarity/drug effects , Diet, High-Fat , Fatty Liver/metabolism , Fatty Liver/pathology , Glucose Intolerance , Homeostasis/drug effects , Inflammation/metabolism , Inflammation/pathology , Interleukin-4/pharmacology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Models, Biological , Nerve Tissue Proteins/deficiency , Phenotype , Protein Binding
10.
Annu Rev Physiol ; 78: 155-80, 2016.
Article in English | MEDLINE | ID: mdl-26667074

ABSTRACT

Glucocorticoid hormones (GC) regulate essential physiological functions including energy homeostasis, embryonic and postembryonic development, and the stress response. From the biomedical perspective, GC have garnered a tremendous amount of attention as highly potent anti-inflammatory and immunosuppressive medications indispensable in the clinic. GC signal through the GC receptor (GR), a ligand-dependent transcription factor whose structure, DNA binding, and the molecular partners that it employs to regulate transcription have been under intense investigation for decades. In particular, next-generation sequencing-based approaches have revolutionized the field by introducing a unified platform for a simultaneous genome-wide analysis of cellular activities at the level of RNA production, binding of transcription factors to DNA and RNA, and chromatin landscape and topology. Here we describe fundamental concepts of GC/GR function as established through traditional molecular and in vivo approaches and focus on the novel insights of GC biology that have emerged over the last 10 years from the rapidly expanding arsenal of system-wide genomic methodologies.


Subject(s)
Genome/genetics , Glucocorticoids/genetics , Glucocorticoids/metabolism , Signal Transduction/physiology , Animals , Genomics/methods , Humans , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
BMC Genomics ; 15: 656, 2014 Aug 06.
Article in English | MEDLINE | ID: mdl-25099603

ABSTRACT

BACKGROUND: Inflammation triggered by infection or injury is tightly controlled by glucocorticoid hormones which signal via a dedicated transcription factor, the Glucocorticoid Receptor (GR), to regulate hundreds of genes. However, the hierarchy of transcriptional responses to GR activation and the molecular basis of their oftentimes non-linear dynamics are not understood. RESULTS: We investigated early glucocorticoid-driven transcriptional events in macrophages, a cell type highly responsive to both pro- and anti-inflammatory stimuli. Using whole transcriptome analyses in resting and acutely lipopolysaccharide (LPS)-stimulated macrophages, we show that early GR target genes form dense networks with the majority of control nodes represented by transcription factors. The expression dynamics of several glucocorticoid-responsive genes are consistent with feed forward loops (FFL) and coincide with rapid GR recruitment. Notably, GR binding sites in genes encoding members of the KLF transcription factor family colocalize with KLF binding sites. Moreover, our gene expression, transcription factor binding and computational data are consistent with the existence of the GR-KLF9-KLF2 incoherent FFL. Analysis of LPS-downregulated genes revealed striking enrichment in multimerized Zn-fingers- and KRAB domain-containing proteins known to bind nucleic acids and repress transcription by propagating heterochromatin. This raises an intriguing possibility that an increase in chromatin accessibility in inflammatory macrophages results from broad downregulation of negative chromatin remodelers. CONCLUSIONS: Pro- and anti-inflammatory stimuli alter the expression of a vast array of transcription factors and chromatin remodelers. By regulating multiple transcription factors, which propagate the initial hormonal signal, GR acts as a coordinating hub in anti-inflammatory responses. As several KLFs promote the anti-inflammatory program in macrophages, we propose that GR and KLFs functionally cooperate to curb inflammation.


Subject(s)
Computational Biology , Gene Regulatory Networks , Macrophages/metabolism , Receptors, Glucocorticoid/metabolism , Transcription Factors/metabolism , Animals , Dexamethasone/pharmacology , Down-Regulation/drug effects , Gene Expression Profiling , Gene Regulatory Networks/drug effects , Glucocorticoids/pharmacology , Kinetics , Lipopolysaccharides/pharmacology , Macrophages/cytology , Macrophages/drug effects , Mice , Models, Biological , Signal Transduction/drug effects , Transcriptional Activation/drug effects
12.
Proc Natl Acad Sci U S A ; 105(51): 20185-90, 2008 Dec 23.
Article in English | MEDLINE | ID: mdl-19074285

ABSTRACT

Transcriptional regulators such as the glucocorticoid receptor (GR) recruit multiple cofactors to activate or repress transcription. Although most cofactors are intrinsically bifunctional, little is known about the molecular mechanisms dictating the specific polarity of regulation. Furthermore, chromatin modifications thought to be confined to silent loci appear in actively transcribed genes suggesting that similar enzymatic activities may mediate constitutive and transient chromatin states. GRIP1, a GR ligand-dependent coregulator of the p160 family can potentiate or inhibit transcription but the molecular contexts and mechanisms that enable GRIP1 corepressor activity are poorly understood. In a yeast 2-hybrid screen with GRIP1 repression domain (RD)-containing fragment, we repeatedly isolated the C-terminal region of a SET domain-containing protein subsequently identified as histone H4 lysine 20 trimethyltransferase, Suv4-20h1. We cloned a full-length Suv4-20h1 and dissected its interaction with GRIP1 in yeast, in vitro, and in mammalian cells. Strict nuclear localization and high salt concentration required for Suv4-20h1 extraction were consistent with its tight association with chromatin. Overexpression of Suv4-20h1 in human U2OS and A549 cells expressing integrated and endogenous GR, respectively, antagonized ligand-dependent induction of a subset of GR target genes, whereas Suv4-20h1 siRNA-mediated depletion had a reciprocal effect. Inhibition of GR transactivation required both the GRIP1 interacting region of Suv4-20h1 and its catalytic activity. Thus, Suv4-20h1 known exclusively as a factor involved in constitutive heterochromatin maintenance, actively participates in hormone-dependent transcriptional regulation affecting GR target gene expression in a promoter- and cell type-specific manner.


Subject(s)
Carrier Proteins/metabolism , Gene Expression Regulation , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Glucocorticoid/genetics , Binding Sites , Carrier Proteins/physiology , Cell Line , Cloning, Molecular , Histone-Lysine N-Methyltransferase/physiology , Humans , Nerve Tissue Proteins/physiology , Protein Binding , Protein Structure, Tertiary , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL