Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 171
Filter
1.
J Autoimmun ; 144: 103177, 2024 04.
Article in English | MEDLINE | ID: mdl-38368767

ABSTRACT

Psoriasis (PS) and atopic dermatitis (AD) are common skin inflammatory diseases characterized by hyper-responsive keratinocytes. Although, some cytokines have been suggested to be specific for each disease, other cytokines might be central to both diseases. Here, we show that Tumor necrosis factor superfamily member 14 (TNFSF14), known as LIGHT, is required for experimental PS, similar to its requirement in experimental AD. Mice devoid of LIGHT, or deletion of either of its receptors, lymphotoxin ß receptor (LTßR) and herpesvirus entry mediator (HVEM), in keratinocytes, were protected from developing imiquimod-induced psoriatic features, including epidermal thickening and hyperplasia, and expression of PS-related genes. Correspondingly, in single cell RNA-seq analysis of PS patient biopsies, LTßR transcripts were found strongly expressed with HVEM in keratinocytes, and LIGHT was upregulated in T cells. Similar transcript expression profiles were also seen in AD biopsies, and LTßR deletion in keratinocytes also protected mice from allergen-induced AD features. Moreover, in vitro, LIGHT upregulated a broad spectrum of genes in human keratinocytes that are clinical features of both PS and AD skin lesions. Our data suggest that agents blocking LIGHT activity might be useful for therapeutic intervention in PS as well as in AD.


Subject(s)
Dermatitis, Atopic , Psoriasis , Humans , Mice , Animals , Receptors, Tumor Necrosis Factor, Member 14/genetics , Receptors, Tumor Necrosis Factor, Member 14/metabolism , Dermatitis, Atopic/genetics , Dermatitis, Atopic/metabolism , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism , Keratinocytes/metabolism , Cytokines/metabolism , Psoriasis/genetics , Psoriasis/metabolism , Inflammation/metabolism
3.
Am J Clin Dermatol ; 25(3): 447-461, 2024 May.
Article in English | MEDLINE | ID: mdl-38236520

ABSTRACT

Atopic dermatitis (AD) is a chronic, heterogeneous, inflammatory disease characterized by skin lesions, pruritus, and pain. Patients with moderate-to-severe AD experience chronic symptoms, intensified by unpredictable flares, and often have comorbidities and secondary complications, which can result in significant clinical burden that impacts the patient's overall quality of life. The complex interplay of immune dysregulation and skin barrier disruption drives AD pathogenesis, of which T-cell-dependent inflammation plays a critical role in patients with AD. Despite new targeted therapies, many patients with moderate-to-severe AD fail to achieve or sustain their individual treatment goals and/or may not be suitable for or tolerate these therapies. There remains a need for a novel, efficacious, well-tolerated therapeutic option that can deliver durable benefits across a heterogeneous AD patient population. Expression of OX40 [tumor necrosis factor receptor superfamily, member 4 (TNFRSF4)], a prominent T-cell co-stimulatory molecule, and its ligand [OX40L; tumor necrosis factor superfamily, member 4 (TNFSF4)] is increased in AD. As the OX40 pathway is critical for expansion, differentiation, and survival of effector and memory T cells, its targeting might be a promising therapeutic approach to provide sustained inhibition of pathogenic T cells and associated inflammation and broad disease control. Antibodies against OX40 [rocatinlimab (AMG 451/KHK4083) and telazorlimab (GBR 830)] or OX40L [amlitelimab (KY1005)] have shown promising results in early-phase clinical studies of moderate-to-severe AD, highlighting the importance of OX40 signaling as a new therapeutic target in AD.


Subject(s)
Dermatitis, Atopic , Molecular Targeted Therapy , OX40 Ligand , Receptors, OX40 , Dermatitis, Atopic/immunology , Dermatitis, Atopic/drug therapy , Humans , Receptors, OX40/antagonists & inhibitors , Receptors, OX40/immunology , Receptors, OX40/metabolism , OX40 Ligand/antagonists & inhibitors , OX40 Ligand/metabolism , Severity of Illness Index , Skin/immunology , Skin/pathology , Quality of Life , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Signal Transduction/immunology , Signal Transduction/drug effects , Treatment Outcome
4.
PLoS Pathog ; 20(1): e1011852, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38236791

ABSTRACT

Conventional antiviral memory CD4 T cells typically arise during the first two weeks of acute infection. Unlike most viruses, cytomegalovirus (CMV) exhibits an extended persistent replication phase followed by lifelong latency accompanied with some gene expression. We show that during mouse CMV (MCMV) infection, CD4 T cells recognizing an epitope derived from the viral M09 protein only develop after conventional memory T cells have already peaked and contracted. Ablating these CD4 T cells by mutating the M09 genomic epitope in the MCMV Smith strain, or inducing them by introducing the epitope into the K181 strain, resulted in delayed or enhanced control of viral persistence, respectively. These cells were shown to be unique compared to their conventional memory counterparts; producing higher IFNγ and IL-2 and lower IL-10 levels. RNAseq analyses revealed them to express distinct subsets of effector genes as compared to classical CD4 T cells. Additionally, when M09 cells were induced by epitope vaccination they significantly enhanced protection when compared to conventional CD4 T cells alone. These data show that late-rising CD4 T cells are a unique memory subset with excellent protective capacities that display a development program strongly differing from the majority of memory T cells.


Subject(s)
Cytomegalovirus Infections , Muromegalovirus , Animals , Mice , CD4-Positive T-Lymphocytes , Epitopes , Salivary Glands , CD8-Positive T-Lymphocytes
5.
Allergy ; 79(1): 116-127, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37650473

ABSTRACT

BACKGROUND: Targeting IL-13 is highly efficacious in patients with Th2-biased atopic dermatitis (AD), but inhibition of other inflammatory molecules might also limit disease. We investigated the importance of the TNF family cytokine TNF-like weak inducer of apoptosis (TWEAK; TNFSF12) to keratinocyte dysregulation and the pathogenesis of AD in mice and also tested if blocking TWEAK has a similar therapeutic effect as targeting IL-13. METHODS: Conditional knockout mice lacking Fn14 (TNFRSF12A), the receptor for TWEAK, only in keratinocytes, were repetitively sensitized with house dust mite allergen and analyzed for AD-like skin inflammation. To determine the translational potential, wild-type mice with AD were therapeutically treated with anti-TWEAK and/or anti-IL-13 antibodies, and skin inflammation was assessed. RESULTS: Mice deficient in Fn14 in keratinocytes were resistant to developing maximal clinical features of AD, exhibiting reduced epidermal hyperplasia and dermal thickening, less skin infiltration of immune cells, and downregulated inflammatory gene expression. Moreover, therapeutic neutralization of TWEAK in wild-type mice with AD reduced all of the pathological features to a comparable extent as blocking IL-13. CONCLUSIONS: The activity of TWEAK in keratinocytes contributes to AD development, and neutralizing TWEAK represents a future potential therapeutic option in human AD similar to targeting IL-13.


Subject(s)
Dermatitis, Atopic , Interleukin-13 , Humans , Animals , Mice , Dermatitis, Atopic/drug therapy , TWEAK Receptor/genetics , TWEAK Receptor/metabolism , Inflammation/metabolism , Apoptosis , Mice, Knockout
7.
Front Immunol ; 14: 1228486, 2023.
Article in English | MEDLINE | ID: mdl-37662949

ABSTRACT

Costimulatory receptors on immune cells represent attractive targets for immunotherapy given that these molecules can increase the frequency of individual protective immune cell populations and their longevity, as well as enhance various effector functions. 4-1BB, a member of the TNF receptor superfamily, also known as CD137 and TNFRSF9, is one such molecule that is inducible on several cell types, including T cells and NK cells. Preclinical studies in animal models have validated the notion that stimulating 4-1BB with agonist reagents or its natural ligand could be useful to augment conventional T cell and NK cell immunity to protect against tumor growth and against viral infection. Additionally, stimulating 4-1BB can enhance regulatory T cell function and might be useful in the right context for suppressing autoimmunity. Two human agonist antibodies to 4-1BB have been produced and tested in clinical trials for cancer, with variable results, leading to the production of a wealth of second-generation antibody constructs, including bi- and multi-specifics, with the hope of optimizing activity and selectivity. Here, we review the progress to date in agonism of 4-1BB, discuss the complications in targeting the immune system appropriately to elicit the desired activity, together with challenges in engineering agonists, and highlight the untapped potential of manipulating this molecule in infectious disease and autoimmunity.


Subject(s)
Antibodies , Immunotherapy , Animals , Humans , Autoimmunity , Killer Cells, Natural , Longevity
8.
J Vis Exp ; (192)2023 02 24.
Article in English | MEDLINE | ID: mdl-36912535

ABSTRACT

Fms-like tyrosine kinase 3 ligand (Flt3L) is a hematopoietic cytokine that promotes the survival and differentiation of dendritic cells (DCs). It has been used in tumor vaccines to activate innate immunity and enhance antitumor responses. This protocol demonstrates a therapeutic model using cell-based tumor vaccine consisting of Flt3L-expressing B16-F10 melanoma cells along with phenotypic and functional analysis of immune cells in the tumor microenvironment (TME). Procedures for cultured tumor cell preparation, tumor implantation, cell irradiation, tumor size measurement, intratumoral immune cell isolation, and flow cytometry analysis are described. The overall goal of this protocol is to provide a preclinical solid tumor immunotherapy model, and a research platform to study the relationship between tumor cells and infiltrating immune cells. The immunotherapy protocol described here can be combined with other therapeutic modalities, such as immune checkpoint blockade (anti-CTLA-4, anti-PD-1, anti-PD-L1 antibodies) or chemotherapy in order to improve the cancer therapeutic effect of melanoma.


Subject(s)
Cancer Vaccines , Melanoma, Experimental , Animals , Humans , Melanoma, Experimental/therapy , Dendritic Cells , Immunotherapy/methods , Cytokines , Vaccination , Tumor Microenvironment
9.
Allergy ; 78(8): 2168-2180, 2023 08.
Article in English | MEDLINE | ID: mdl-36951658

ABSTRACT

BACKGROUND: Exacerbations of asthma are thought to be strongly dependent on reactivation of allergen-induced lung tissue-resident and circulatory memory CD4 T cells. Strategies that broadly inhibit multiple T cell populations might then be useful to limit asthma. Accordingly, we tested whether targeting CD3 during exposure to inhaled allergen could prevent the accumulation of lung-localized effector memory CD4 T cells and block exacerbations of asthmatic inflammation. METHODS: House dust mite-sensitized and repetitively challenged BL/6 mice were transiently treated therapeutically with F(ab')2 anti-CD3ε and memory T cell responses and lung inflammation were assessed. PBMCs from HDM-allergic donors were examined for the effect of anti-CD3 on expansion of allergen-reactive T cells. RESULTS: Allergen-sensitized mice undergoing exacerbations of asthma were protected from lung inflammation by transient therapeutic treatment with F(ab')2 anti-CD3. Regardless of whether sensitized mice underwent a secondary or tertiary recall response to inhaled allergen, anti-CD3 inhibited all phenotypes of effector memory CD4 T cells in the lung tissue and lung vasculature by 80%-90%, including those derived from tissue-resident and circulatory memory T cells. This did not depend on Treg cells suggesting it was primarily a blocking effect on memory T cell signaling. Correspondingly, anti-CD3 also strongly inhibited proliferation of human allergen-reactive memory CD4 T cells from allergic individuals. In contrast, the number of surviving tissue-resident memory CD4 T cells that were maintained in the lungs at later times was not robustly reduced by anti-CD3. CONCLUSION: Anti-CD3 F(ab')2 administration at the time of allergen exposure represents a viable strategy for limiting the immediate activity of allergen-responding memory T cells and asthma exacerbations.


Subject(s)
Asthma , Hypersensitivity , Pneumonia , Animals , Mice , Humans , Memory T Cells , CD4-Positive T-Lymphocytes , Th2 Cells , Asthma/prevention & control , Allergens/adverse effects , Pyroglyphidae , Disease Models, Animal
10.
J Allergy Clin Immunol ; 151(4): 976-990.e5, 2023 04.
Article in English | MEDLINE | ID: mdl-36473503

ABSTRACT

BACKGROUND: Dysregulation of airway smooth muscle cells (ASM) is central to the severity of asthma. Which molecules dominantly control ASM in asthma is unclear. High levels of the cytokine LIGHT (aka TNFSF14) have been linked to asthma severity and lower baseline predicted FEV1 percentage, implying that signals through its receptors might directly control ASM dysfunction. OBJECTIVE: Our study sought to determine whether signaling via lymphotoxin beta receptor (LTßR) or herpesvirus entry mediator from LIGHT dominantly drives ASM hyperreactivity induced by allergen. METHODS: Conditional knockout mice deficient for LTßR or herpesvirus entry mediator in smooth muscle cells were used to determine their role in ASM deregulation and airway hyperresponsiveness (AHR) in vivo. Human ASM were used to study signals induced by LTßR. RESULTS: LTßR was strongly expressed in ASM from normal and asthmatic subjects compared to several other receptors implicated in smooth muscle deregulation. Correspondingly, conditional deletion of LTßR only in smooth muscle cells in smMHCCreLTßRfl/fl mice minimized changes in their numbers and mass as well as AHR induced by house dust mite allergen in a model of severe asthma. Intratracheal LIGHT administration independently induced ASM hypertrophy and AHR in vivo dependent on direct LTßR signals to ASM. LIGHT promoted contractility, hypertrophy, and hyperplasia of human ASM in vitro. Distinguishing LTßR from the receptors for IL-13, TNF, and IL-17, which have also been implicated in smooth muscle dysregulation, LIGHT promoted NF-κB-inducing kinase-dependent noncanonical nuclear factor kappa-light-chain enhancer of activated B cells in ASM in vitro, leading to sustained accumulation of F-actin, phosphorylation of myosin light chain kinase, and contractile activity. CONCLUSIONS: LTßR signals directly and dominantly drive airway smooth muscle hyperresponsiveness relevant for pathogenesis of airway remodeling in severe asthma.


Subject(s)
Asthma , Receptors, Tumor Necrosis Factor, Member 14 , Humans , Mice , Animals , Lymphotoxin beta Receptor/genetics , Asthma/pathology , Muscle, Smooth , Myocytes, Smooth Muscle/pathology , Mice, Knockout , Allergens , Lung/pathology
11.
J Immunol ; 2022 Oct 26.
Article in English | MEDLINE | ID: mdl-36426994

ABSTRACT

Eosinophilic esophagitis (EoE) is a chronic type 2 allergic disease, with esophageal tissue remodeling as the mechanism behind clinical dysphagia and strictures. IL-13 is thought to be a central driver of disease, but other inflammatory factors, such as IFNs and TNF superfamily members, have been hypothesized to play a role in disease pathogenesis. We recently found that the cytokine TNFSF14/LIGHT is upregulated in the esophagus of patients with EoE and that LIGHT promotes inflammatory activity in esophageal fibroblasts. However, the global effects of LIGHT on EoE pathogenesis in vivo remain unknown. We investigated the impact of a LIGHT deficiency in a murine model of EoE driven by house dust mite allergen. Chronic intranasal challenge with house dust mite promoted esophageal eosinophilia and increased CD4+ T cell numbers and IL-13 and CCL11 production in wild-type mice. Esophageal remodeling was reflected by submucosal collagen accumulation, increased muscle density, and greater numbers of fibroblasts. LIGHT-/- mice displayed normal esophageal eosinophilia, but exhibited reduced frequencies of CD4 T cells, IL-13 expression, submucosal collagen, and muscle density and a decrease in esophageal accumulation of fibroblasts. In vitro, LIGHT increased division of human esophageal fibroblasts and selectively enhanced IL-13-mediated expression of a subset of inflammatory and fibrotic genes. These results show that LIGHT contributes to various features of murine EoE, impacting the accumulation of CD4 T cells, IL-13 production, fibroblast proliferation, and esophagus remodeling. These findings suggest that LIGHT may be, to our knowledge, a novel therapeutic target for the treatment of EoE.

12.
J Immunol ; 2022 Oct 26.
Article in English | MEDLINE | ID: mdl-36288906

ABSTRACT

Eosinophilic esophagitis (EoE) is a chronic type 2 allergic disease, with esophageal tissue remodeling as the mechanism behind clinical dysphagia and strictures. IL-13 is thought to be a central driver of disease, but other inflammatory factors, such as IFNs and TNF superfamily members, have been hypothesized to play a role in disease pathogenesis. We recently found that the cytokine TNFSF14/LIGHT is upregulated in the esophagus of patients with EoE and that LIGHT promotes inflammatory activity in esophageal fibroblasts. However, the global effects of LIGHT on EoE pathogenesis in vivo remain unknown. We investigated the impact of a LIGHT deficiency in a murine model of EoE driven by house dust mite allergen. Chronic intranasal challenge with house dust mite promoted esophageal eosinophilia and increased CD4+ T cell numbers and IL-13 and CCL11 production in wild-type mice. Esophageal remodeling was reflected by submucosal collagen accumulation, increased muscle density, and greater numbers of fibroblasts. LIGHT-/- mice displayed normal esophageal eosinophilia, but exhibited reduced frequencies of CD4 T cells, IL-13 expression, submucosal collagen, and muscle density and a decrease in esophageal accumulation of fibroblasts. In vitro, LIGHT increased division of human esophageal fibroblasts and selectively enhanced IL-13-mediated expression of a subset of inflammatory and fibrotic genes. These results show that LIGHT contributes to various features of murine EoE, impacting the accumulation of CD4 T cells, IL-13 production, fibroblast proliferation, and esophagus remodeling. These findings suggest that LIGHT may be, to our knowledge, a novel therapeutic target for the treatment of EoE.

13.
Front Immunol ; 13: 951361, 2022.
Article in English | MEDLINE | ID: mdl-35936001

ABSTRACT

Tissue-resident memory CD4 T cells (Trm) are thought to be a major contributor to asthma relapse, but the role of circulatory T cells in asthma exacerbations or to maintaining the population of lung Trm cells is not fully understood. Here, we used a house dust mite allergen-based murine model of asthma relapse, and monitored the development of lung effector/Trm phenotype CD44hiCD62LloCD69+ CD4 T cells. To determine the contribution of circulatory cells, mice were treated with FTY720, to block lymphocyte egress from lymph nodes. Inhibiting the primary migration of circulatory cells to the lungs mitigated the accumulation and expansion of allergen-driven Trm phenotype cells, but subsequent allergen challenges still resulted in strong lung inflammation and Trm cell accumulation. This was blocked if FTY720 was also given at the time of allergen re-exposure, showing that new circulatory cells contributed to this lung memory/effector T cell pool at times well after the initial sensitization. However, once lung-localized Trm cells developed at high frequency, circulatory cells were not required to maintain this population following allergen re-encounter, even though circulatory cells still were major contributors to the overall asthmatic lung inflammatory response. Our results suggest that strategies that target the response of circulatory memory T cells and Trm cells together might be required to strongly inhibit T cell reactivity to airborne allergens and to limit exacerbations of asthma and their reoccurrence, but the contribution of circulatory T cells might vary in long-term asthmatics possessing a large stable Trm cell population in the lungs.


Subject(s)
Asthma , Pneumonia , Allergens , Animals , Asthma/pathology , CD4-Positive T-Lymphocytes , Fingolimod Hydrochloride/pharmacology , Immunologic Memory , Lung , Mice , Pneumonia/pathology , Recurrence
14.
J Exp Med ; 219(7)2022 07 04.
Article in English | MEDLINE | ID: mdl-35604387

ABSTRACT

Advances in understanding the physiologic functions of the tumor necrosis factor superfamily (TNFSF) of ligands, receptors, and signaling networks are providing deeper insight into pathogenesis of infectious and autoimmune diseases and cancer. LIGHT (TNFSF14) has emerged as an important modulator of critical innate and adaptive immune responses. LIGHT and its signaling receptors, herpesvirus entry mediator (TNFRSF14), and lymphotoxin ß receptor, form an immune regulatory network with two co-receptors of herpesvirus entry mediator, checkpoint inhibitor B and T lymphocyte attenuator, and CD160. Deciphering the fundamental features of this network reveals new understanding to guide therapeutic development. Accumulating evidence from infectious diseases points to the dysregulation of the LIGHT network as a disease-driving mechanism in autoimmune and inflammatory reactions in barrier organs, including coronavirus disease 2019 pneumonia and inflammatory bowel diseases. Recent clinical results warrant further investigation of the LIGHT regulatory network and application of target-modifying therapeutics for disease intervention.


Subject(s)
COVID-19 , Receptors, Tumor Necrosis Factor, Member 14 , Humans , Inflammation , Signal Transduction , T-Lymphocytes
15.
Mucosal Immunol ; 15(2): 327-337, 2022 02.
Article in English | MEDLINE | ID: mdl-34903876

ABSTRACT

Fibroblasts mediate tissue remodeling in eosinophilic esophagitis (EoE), a chronic allergen-driven inflammatory pathology. Diverse fibroblast subtypes with homeostasis-regulating or inflammatory profiles have been recognized in various tissues, but which mediators induce these alternate differentiation states remain largely unknown. We recently identified that TNFSF14/LIGHT promotes an inflammatory esophageal fibroblast in vitro. Herein we used esophageal biopsies and primary fibroblasts to investigate the role of the LIGHT receptors, herpes virus entry mediator (HVEM) and lymphotoxin-beta receptor (LTßR), and their downstream activated pathways, in EoE. In addition to promoting inflammatory gene expression, LIGHT down-regulated homeostatic factors including WNTs, BMPs and type 3 semaphorins. In vivo, WNT2B+ fibroblasts were decreased while ICAM-1+ and IL-34+ fibroblasts were expanded in EoE, suggesting that a LIGHT-driven gene signature was imprinted in EoE versus normal esophageal fibroblasts. HVEM and LTßR overexpression and deficiency experiments demonstrated that HVEM regulates a limited subset of LIGHT targets, whereas LTßR controls all transcriptional effects. Pharmacologic blockade of the non-canonical NIK/p100/p52-mediated NF-κB pathway potently silenced LIGHT's transcriptional effects, with a lesser role found for p65 canonical NF-κB. Collectively, our results show that LIGHT promotes differentiation of esophageal fibroblasts toward an inflammatory phenotype and represses homeostatic gene expression via a LTßR-NIK-p52 NF-κB dominant pathway.


Subject(s)
Esophagus , Inflammation , Transcriptome , Tumor Necrosis Factor Ligand Superfamily Member 14 , Esophagus/metabolism , Fibroblasts/metabolism , Homeostasis , Humans , Inflammation/genetics , Lymphotoxin beta Receptor/genetics , Lymphotoxin beta Receptor/metabolism , NF-kappa B/metabolism , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 14/genetics , Tumor Necrosis Factor Ligand Superfamily Member 14/metabolism
16.
J Med Imaging Radiat Oncol ; 66(6): 768-775, 2022 Sep.
Article in English | MEDLINE | ID: mdl-34799981

ABSTRACT

INTRODUCTION: To optimize CT chest protocol by comparing venous contrast timing with arterial timing for contrast opacification in vessels, qualitative image quality and radiologists' satisfaction and diagnostic confidence in assessing for potential nodal, pleural and pulmonary disease in general oncology outpatients. METHOD: Matched case-control study performed following CT protocol update. 92 patients with a range of primary malignancies with 2 CT chests in a 2-year period, one with an arterial phase protocol and the second in the 60 second venous phase, were included. Contrast attenuation in aorta, pulmonary artery and liver were measured. Subjective measurements assessed perivenous artefact, confidence in nodal pleural and pulmonary assessment and presence of pulmonary emboli. Statistical analysis was performed using paired and unpaired t-tests. RESULTS: Venous-phase CT demonstrated more consistent enhancement of the vessels, with higher attenuation of the nodes, pulmonary and pleural lesions. There was a significant reduction in perivenous beam hardening artefact on venous-phase CT (P < 0.001). Diagnostic confidence was significantly higher for nodal assessment and pleural abnormality visibility (P < 0.001) and pleural assessment (P < 0.05). There was no significant difference in pulmonary mass visibility. There was adequate enhancement to diagnose significant pulmonary emboli (PE) with 4 incidental PEs detected on the venous phase, extending to segmental vessels. CONCLUSION: Venous-phase CT chest performs better than arterial-phase on all fronts, without compromising assessment of incidental pulmonary emboli. When intravenous contrast is indicated in a routine chest CT (excluding a CT-angiogram), the default timing should be a venous or 60s phase.


Subject(s)
Contrast Media , Pulmonary Embolism , Angiography/methods , Case-Control Studies , Humans , Pulmonary Embolism/diagnostic imaging , Thorax , Tomography, X-Ray Computed/methods
17.
Radiol Res Pract ; 2021: 7191348, 2021.
Article in English | MEDLINE | ID: mdl-34853706

ABSTRACT

INTRODUCTION: This manuscript aims to investigate the amount of intraluminal gas in acute, nonperforated appendicitis identified on computed tomography (CT) in diagnosing gangrenous appendicitis. METHODS: This is a retrospective observational, case-control study with consecutive data collected at a tertiary institution over a two-year period, of patients with CT-diagnosed acute appendicitis who subsequently went on for surgery within 48 hours. Patients who were less than 16 years old, who had an interval between CT and surgery of more than 48 hours, or with CT evidence of appendiceal perforation were excluded. Images were independently assessed by 3 radiologists for intraluminal gas, and the results were then correlated with reference standards obtained from surgical and histopathology reports for the diagnosis of nongangrenous versus gangrenous appendicitis. The sensitivity, specificity, and predictive values of CT intraluminal gas in gangrenous appendicitis were calculated. RESULTS: Our study identified 93 patients with nonperforated acute appendicitis who underwent surgery within the stated timeframe. Intraluminal gas in the appendix was identified in 26 patients (28%), of which 54% had macroscopic and/or microscopic evidence of gangrenous appendicitis. This is in contrast to the subgroup of patients who did not have intraluminal gas (72%), of which only 33% had gangrenous appendicitis. The specificity of intraluminal gas for gangrenous appendicitis is 79%, with a negative predictive value of 86% and likelihood ratio of 1.85. CONCLUSION: In cases of established acute appendicitis, the presence of intraluminal gas is a moderately specific sign for gangrenous complication. This is worth reporting as it can help prognosticate and triage patients accordingly, for a timelier surgical management and a better outcome.

18.
Sci Immunol ; 6(65): eabi8823, 2021 Nov 19.
Article in English | MEDLINE | ID: mdl-34797693

ABSTRACT

TNF and IL-17 are two cytokines that drive dysregulated keratinocyte activity, and their targeting is highly efficacious in patients with psoriasis, but whether these molecules act with other inflammatory factors is not clear. Here, we show that mice having a keratinocyte-specific deletion of Fn14 (Tnfrsf12a), the receptor for the TNF superfamily cytokine TWEAK (Tnfsf12), displayed reduced imiquimod-induced skin inflammation, including diminished epidermal hyperplasia and less expression of psoriasis signature genes. This corresponded with Fn14 being expressed in keratinocytes in human psoriasis lesions and TWEAK being found in several subsets of skin cells. Transcriptomic studies in human keratinocytes revealed that TWEAK strongly overlaps with IL-17A and TNF in up-regulating the expression of CXC chemokines, along with cytokines such as IL-23 and inflammation-associated proteins like S100A8/9 and SERPINB1/B9, all previously found to be highly expressed in the lesional skin of patients with psoriasis. TWEAK displayed strong synergism with TNF or IL-17A in up-regulating messenger RNA for many psoriasis-associated genes in human keratinocytes, including IL23A, IL36G, and multiple chemokines, implying that TWEAK acts with TNF and IL-17 to enhance feedback inflammatory activity. Correspondingly, therapeutic treatment of mice with anti-TWEAK was equally as effective as antibodies to IL-17A or TNF in reducing clinical and immunological features of psoriasis-like skin inflammation and combination targeting of TWEAK with either cytokine had no greater inhibitory effect, reinforcing the conclusion that all three cytokines function together. Thus, blocking TWEAK could be comparable to targeting TNF or IL-17 and might be considered as an alternate therapeutic treatment for psoriasis.


Subject(s)
Cytokine TWEAK/immunology , Interleukin-17/immunology , Keratinocytes/immunology , Psoriasis/immunology , Tumor Necrosis Factors/immunology , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Psoriasis/therapy
19.
Korean J Radiol ; 22(12): 2006-2016, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34564959

ABSTRACT

The expanding scope of interventional musculoskeletal procedures has resulted in increased pressure on general radiologists. The confidence of general radiologists in performing ultrasound-guided musculoskeletal procedures varies with their clinical exposure. This didactic review provides a methodologically and clinically oriented approach to enhancing user understanding and confidence in performing ultrasound-guided musculoskeletal procedures. The body of the text is accompanied by figures depicting the procedural approach, injection site, and labeled ultrasonography images. This paper aims to provide a teaching and bedside aid for education on and the execution of musculoskeletal procedures to ensure the provision of quality health care.


Subject(s)
Radiologists , Ultrasonography, Interventional , Humans , Toes , Ultrasonography
20.
J Allergy Clin Immunol ; 147(3): 1087-1092.e3, 2021 03.
Article in English | MEDLINE | ID: mdl-33065121

ABSTRACT

BACKGROUND: Poor clearance of apoptotic cells has been suggested to contribute to severe asthma, but whether uptake of apoptotic cells by lung phagocytes might dampen house dust mite (HDM)-induced lung inflammation has not been shown. OBJECTIVES: This study investigated whether apoptotic cell engulfment in the murine lung impacts the development of allergen-induced asthmatic airway inflammation and which immune modulating mechanisms were activated. METHODS: Apoptotic cells were infused into the lungs of mice challenged with HDM allergen and lung inflammation, expression of suppressive molecules, and induction of regulatory T cells were monitored. Additionally, an adenosine receptor agonist was tested to study the mechanism of suppression elicited by apoptotic cells. RESULTS: Apoptotic cell uptake by lung alveolar macrophages suppressed HDM-driven allergic asthma. This was associated with promoting the regulatory T cell-inducing molecule retinoic acid, inhibiting inflammatory cytokine production, and making macrophages more susceptible to receiving suppressive signals from adenosine. Correspondingly, adenosine receptor agonist treatment also limited HDM-driven allergic airway inflammation through an action on alveolar macrophages. CONCLUSIONS: These data provide insight into the mechanisms by which lung macrophages dampen allergen-induced airway inflammation. They suggest that targeting lung macrophages to increase their phagocytic capacity, enhance their ability to make retinoic acid, dampen their capacity to make inflammatory cytokines, and increase their responsiveness to adenosine, could be useful to suppress allergic responses.


Subject(s)
Asthma/immunology , Hypersensitivity/immunology , Macrophages, Alveolar/immunology , Respiratory Hypersensitivity/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, Dermatophagoides/immunology , Apoptosis , Cells, Cultured , Disease Models, Animal , Humans , Male , Mice , Mice, Inbred C57BL , Pyroglyphidae
SELECTION OF CITATIONS
SEARCH DETAIL
...