Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Leuk Lymphoma ; : 1-11, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38861618

ABSTRACT

We aim to investigate cardiovascular mortality risk among diffuse large B-cell lymphoma (DLBCL) patients and explore cardiovascular mortality trends in the past decades in United States. We extracted data from the Surveillance, Epidemiology, and End Results database for adult patients diagnosed with DLBCL between 1975 and 2019. Standardized mortality ratio, joinpoint regression analysis, and competing risk model were analyzed. Overall, 49,918 patients were enrolled, of whom 4167 (8.3%) cardiovascular deaths were observed, which was 1.22 times the number expected (95%CI, 1.19-1.26). During 1985-2019, the incidence-based cardiovascular mortality rate increased by 0.98% per year (95%CI, 0.58-1.39%), with statistically significant increases in age groups younger than 75 years. The cumulative mortality from cardiovascular disease increased by age but never exceeded that from DLBCL. Older age, male sex, earlier year of diagnosis, lower tumor stage at diagnosis, chemotherapy, radiotherapy, and surgery were all poor prognostic factors for cardiovascular mortality.

2.
Int J Biol Sci ; 20(6): 2092-2110, 2024.
Article in English | MEDLINE | ID: mdl-38617538

ABSTRACT

Development of non-surgical treatment of human abdominal aortic aneurysm (AAA) has clinical significance. Colchicine emerges as an effective therapeutic regimen in cardiovascular diseases. Yet, whether colchicine slows AAA growth remain controversy. Here, we demonstrated that daily intragastric administration of low-dose colchicine blocked AAA formation, prevented vascular smooth muscle cell (SMC) phenotype switching and apoptosis, and vascular inflammation in both peri-aortic CaPO4 injury and subcutaneous angiotensin-II infusion induced experimental AAA mice models. Mechanistically, colchicine increased global mRNA stability by inhibiting the METTL14/YTHDC1-mediated m6A modification, resulting in increased sclerostin (SOST) expression and consequent inactivation of the WNT/ß-catenin signaling pathway in vascular SMCs from mouse AAA lesions and in cultured human aortic SMCs. Moreover, human and mouse AAA lesions all showed increased m6A methylation, decreased SOST expression, and skewed synthetic SMC de-differentiation phenotype, compared to those without AAA. This study uncovers a novel mechanism of colchicine in slowing AAA development by using the METTL14/SOST/WNT/ß-catenin axis to control vascular SMC homeostasis in mouse aortic vessels and in human aortic SMCs. Therefore, use of colchicine may benefit AAA patients in clinical practice.


Subject(s)
Aortic Aneurysm, Abdominal , Muscle, Smooth, Vascular , Humans , Animals , Mice , Aortic Aneurysm, Abdominal/drug therapy , Homeostasis , Aorta , Colchicine/therapeutic use
3.
Eur Heart J ; 44(29): 2763-2783, 2023 08 01.
Article in English | MEDLINE | ID: mdl-37279475

ABSTRACT

AIMS: Blood eosinophil count and eosinophil cationic protein (ECP) concentration are risk factors of cardiovascular diseases. This study tested whether and how eosinophils and ECP contribute to vascular calcification and atherogenesis. METHODS AND RESULTS: Immunostaining revealed eosinophil accumulation in human and mouse atherosclerotic lesions. Eosinophil deficiency in ΔdblGATA mice slowed atherogenesis with increased lesion smooth muscle cell (SMC) content and reduced calcification. This protection in ΔdblGATA mice was muted when mice received donor eosinophils from wild-type (WT), Il4-/-, and Il13-/- mice or mouse eosinophil-associated-ribonuclease-1 (mEar1), a murine homologue of ECP. Eosinophils or mEar1 but not interleukin (IL) 4 or IL13 increased the calcification of SMC from WT mice but not those from Runt-related transcription factor-2 (Runx2) knockout mice. Immunoblot analyses showed that eosinophils and mEar1 activated Smad-1/5/8 but did not affect Smad-2/3 activation or expression of bone morphogenetic protein receptors (BMPR-1A/1B/2) or transforming growth factor (TGF)-ß receptors (TGFBR1/2) in SMC from WT and Runx2 knockout mice. Immunoprecipitation showed that mEar1 formed immune complexes with BMPR-1A/1B but not TGFBR1/2. Immunofluorescence double-staining, ligand binding, and Scatchard plot analysis demonstrated that mEar1 bound to BMPR-1A and BMPR-1B with similar affinity. Likewise, human ECP and eosinophil-derived neurotoxin (EDN) also bound to BMPR-1A/1B on human vascular SMC and promoted SMC osteogenic differentiation. In a cohort of 5864 men from the Danish Cardiovascular Screening trial and its subpopulation of 394 participants, blood eosinophil counts and ECP levels correlated with the calcification scores of different arterial segments from coronary arteries to iliac arteries. CONCLUSION: Eosinophils release cationic proteins that can promote SMC calcification and atherogenesis using the BMPR-1A/1B-Smad-1/5/8-Runx2 signalling pathway.


Subject(s)
Atherosclerosis , Vascular Calcification , Male , Humans , Animals , Mice , Eosinophils , Core Binding Factor Alpha 1 Subunit/metabolism , Blood Proteins/analysis , Osteogenesis , Bone Morphogenetic Protein Receptors/metabolism , Interleukin-13/metabolism , Eosinophil Granule Proteins/metabolism , Ribonucleases/metabolism , Atherosclerosis/metabolism , Mice, Knockout
4.
Cardiovasc Res ; 119(1): 195-212, 2023 03 17.
Article in English | MEDLINE | ID: mdl-35394031

ABSTRACT

AIMS: Blood eosinophil (EOS) counts and EOS cationic protein (ECP) levels associate positively with major cardiovascular disease (CVD) risk factors and prevalence. This study investigates the role of EOS in cardiac hypertrophy. METHODS AND RESULTS: A retrospective cross-section study of 644 consecutive inpatients with hypertension examined the association between blood EOS counts and cardiac hypertrophy. Pressure overload- and ß-adrenoreceptor agonist isoproterenol-induced cardiac hypertrophy was produced in EOS-deficient ΔdblGATA mice. This study revealed positive correlations between blood EOS counts and left ventricular (LV) mass and mass index in humans. ΔdblGATA mice showed exacerbated cardiac hypertrophy and dysfunction, with increased LV wall thickness, reduced LV internal diameter, and increased myocardial cell size, death, and fibrosis. Repopulation of EOS from wild-type (WT) mice, but not those from IL4-deficient mice ameliorated cardiac hypertrophy and cardiac dysfunctions. In ΔdblGATA and WT mice, administration of ECP mEar1 improved cardiac hypertrophy and function. Mechanistic studies demonstrated that EOS expression of IL4, IL13, and mEar1 was essential to control mouse cardiomyocyte hypertrophy and death and cardiac fibroblast TGF-ß signalling and fibrotic protein synthesis. The use of human cardiac cells yielded the same results. Human ECP, EOS-derived neurotoxin, human EOS, or murine recombinant mEar1 reduced human cardiomyocyte death and hypertrophy and human cardiac fibroblast TGF-ß signalling. CONCLUSION: Although blood EOS counts correlated positively with LV mass or LV mass index in humans, this study established a cardioprotective role for EOS IL4 and cationic proteins in cardiac hypertrophy and tested a therapeutic possibility of ECPs in this human CVD.


Subject(s)
Eosinophils , Hypertrophy, Left Ventricular , Mice , Humans , Animals , Hypertrophy, Left Ventricular/chemically induced , Hypertrophy, Left Ventricular/prevention & control , Eosinophils/metabolism , Retrospective Studies , Interleukin-4/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/prevention & control , Myocytes, Cardiac/metabolism , Adrenergic beta-Agonists/pharmacology , Transforming Growth Factor beta/metabolism , Fibrosis , Ventricular Remodeling
5.
Microbiol Spectr ; 10(4): e0106822, 2022 08 31.
Article in English | MEDLINE | ID: mdl-35862975

ABSTRACT

The tumor microbiome is believed to have a profound impact on tumor progression owing to its local colonization in the tumor microenvironment (TME). Using the Cancer Microbiome Atlas (TCMA), a database of curated, decontaminated microbial profiles for 3,689 oropharyngeal, esophageal, gastrointestinal, and colorectal tissue samples from 1,772 patients, we conducted a comprehensive multi-omics analysis to reveal microbial signatures among various cancers and the potential mechanisms involved in tumor progression of head and neck squamous cell carcinoma (HNSC). We found that compared with other cancer types, the tumor-resident microbiome of HNSC accounted for the highest bacterial abundance and strongest association with host TME signatures. Fusobacterium was found to be enriched in HNSC tissues, which was associated with an increased inflammatory effect and inferior prognosis. Moreover, we revealed that the microbiota-associated inflammatory TME was attributed to the competing endogenouse RNA (ceRNA) network and chromatin accessibility. IMPORTANCE Studies on revealing the composition and potential mechanisms of the tumor microbiome are still at an initial stage. We uncovered the potential contribution of the tumor-resident microbiota on the immunosuppressive microenvironment in HNSC, which will provide a new perspective for tumor microbiome research and yield valuable insights into the clinical management of HNSC.


Subject(s)
Head and Neck Neoplasms , Fusobacterium , Humans , Prognosis , Squamous Cell Carcinoma of Head and Neck , Tumor Microenvironment/genetics
6.
Clinics (Sao Paulo) ; 76: e2690, 2021.
Article in English | MEDLINE | ID: mdl-34231707

ABSTRACT

This study aimed to explore the relationship between plasma interleukin 6 (IL-6) levels, adverse cardiovascular events, and the severity of acute coronary syndrome (ACS). A literature review was performed of studies regarding IL-6 and ACS extracted from databases including EMBASE, Cqvip, MEDLINE, Web of Knowledge, PubMed, Cochrane Library, China National Knowledge Infrastructure, and Wanfang data. The Newcastle-Ottawa scale (NOS) was used to evaluate the quality of the literature. The literature was screened, its quality was evaluated, and relevant data were extracted for performing meta-analysis using RevMan software (version 5.3). A total of 524 studies were included in the initial survey. After several rounds of screening and analysis, six studies met the inclusion criteria and underwent meta-analysis using a fixed-effect model. Patients were divided into non-severe and severe groups based on the concentration of high-sensitivity C-reactive protein. Meta-analysis of the relationship between IL-6 and the severity of ACS showed that the plasma IL-6 level of patients in the severe group was significantly higher than that of patients in the non-severe group (p<0.00001). Additionally, patients with experience of major adverse cardiovascular events had significantly higher plasma IL-6 levels than did patients without experience of such events (p<0.00001). In summary, patients with ACS and high IL-6 levels tended to be in a critical condition, with a higher risk of adverse cardiovascular events and worse prognosis. Thus, IL-6 levels could indicate whether patients with ACS may have adverse cardiovascular events and determine the severity of ACS.


Subject(s)
Acute Coronary Syndrome , Interleukin-6 , C-Reactive Protein , China , Humans , Prognosis
8.
Circ Res ; 128(2): 188-202, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33153394

ABSTRACT

RATIONALE: Blood eosinophil count and ECP (eosinophil cationic protein) associate with human cardiovascular diseases. Yet, whether eosinophils play a role in cardiovascular disease remains untested. The current study detected eosinophil accumulation in human and murine abdominal aortic aneurysm (AAA) lesions, suggesting eosinophil participation in this aortic disease. OBJECTIVE: To test whether and how eosinophils affect AAA growth. METHODS AND RESULTS: Population-based randomized clinically controlled screening trials revealed higher blood eosinophil count in 579 male patients with AAA than in 5063 non-AAA control (0.236±0.182 versus 0.211±0.154, 109/L, P<0.001). Univariate (odds ratio, 1.381, P<0.001) and multivariate (odds ratio, 1.237, P=0.031) logistic regression analyses indicated that increased blood eosinophil count in patients with AAA served as an independent risk factor of human AAA. Immunostaining and immunoblot analyses detected eosinophil accumulation and eosinophil cationic protein expression in human and murine AAA lesions. Results showed that eosinophil deficiency exacerbated AAA growth with increased lesion inflammatory cell contents, matrix-degrading protease activity, angiogenesis, cell proliferation and apoptosis, and smooth muscle cell loss using angiotensin-II perfusion-induced AAA in Apoe-/- and eosinophil-deficient Apoe-/-ΔdblGATA mice. Eosinophil deficiency increased lesion chemokine expression, muted lesion expression of IL (interleukin) 4 and eosinophil-associated-ribonuclease-1 (mEar1 [mouse EOS-associated-ribonuclease-1], human ECP homolog), and slanted M1 macrophage polarization. In cultured macrophages and monocytes, eosinophil-derived IL4 and mEar1 polarized M2 macrophages, suppressed CD11b+Ly6Chi monocytes, and increased CD11b+Ly6Clo monocytes. mEar1 treatment or adoptive transfer of eosinophil from wild-type and Il13-/- mice, but not eosinophil from Il4-/- mice, blocked AAA growth in Apoe-/-ΔdblGATA mice. Immunofluorescent staining and immunoblot analyses demonstrated a role for eosinophil IL4 and mEar1 in blocking NF-κB (nuclear factor-κB) activation in macrophages, smooth muscle cells, and endothelial cells. CONCLUSIONS: Eosinophils play a protective role in AAA by releasing IL4 and cationic proteins such as mEar1 to regulate macrophage and monocyte polarization and to block NF-κB activation in aortic inflammatory and vascular cells.


Subject(s)
Aorta, Abdominal/metabolism , Aortic Aneurysm, Abdominal/prevention & control , Eosinophils/metabolism , Vascular Remodeling , Adoptive Transfer , Aged , Angiotensin II , Animals , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/metabolism , Aortic Aneurysm, Abdominal/pathology , Cells, Cultured , Dilatation, Pathologic , Disease Models, Animal , Eosinophils/transplantation , Female , Humans , Inflammation Mediators/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Interleukin-4/genetics , Interleukin-4/metabolism , Macrophages/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Monocytes/metabolism , NF-kappa B/metabolism , Phenotype , Ribonucleases/metabolism
9.
Clinics ; 76: e2690, 2021. tab, graf
Article in English | LILACS | ID: biblio-1278911

ABSTRACT

This study aimed to explore the relationship between plasma interleukin 6 (IL-6) levels, adverse cardiovascular events, and the severity of acute coronary syndrome (ACS). A literature review was performed of studies regarding IL-6 and ACS extracted from databases including EMBASE, Cqvip, MEDLINE, Web of Knowledge, PubMed, Cochrane Library, China National Knowledge Infrastructure, and Wanfang data. The Newcastle-Ottawa scale (NOS) was used to evaluate the quality of the literature. The literature was screened, its quality was evaluated, and relevant data were extracted for performing meta-analysis using RevMan software (version 5.3). A total of 524 studies were included in the initial survey. After several rounds of screening and analysis, six studies met the inclusion criteria and underwent meta-analysis using a fixed-effect model. Patients were divided into non-severe and severe groups based on the concentration of high-sensitivity C-reactive protein. Meta-analysis of the relationship between IL-6 and the severity of ACS showed that the plasma IL-6 level of patients in the severe group was significantly higher than that of patients in the non-severe group (p<0.00001). Additionally, patients with experience of major adverse cardiovascular events had significantly higher plasma IL-6 levels than did patients without experience of such events (p<0.00001). In summary, patients with ACS and high IL-6 levels tended to be in a critical condition, with a higher risk of adverse cardiovascular events and worse prognosis. Thus, IL-6 levels could indicate whether patients with ACS may have adverse cardiovascular events and determine the severity of ACS.


Subject(s)
Humans , Interleukin-6 , Acute Coronary Syndrome , Prognosis , C-Reactive Protein , China
10.
Nat Commun ; 11(1): 6396, 2020 12 16.
Article in English | MEDLINE | ID: mdl-33328477

ABSTRACT

Clinical studies reveal changes in blood eosinophil counts and eosinophil cationic proteins that may serve as risk factors for human coronary heart diseases. Here we report an increase of blood or heart eosinophil counts in humans and mice after myocardial infarction (MI), mostly in the infarct region. Genetic or inducible depletion of eosinophils exacerbates cardiac dysfunction, cell death, and fibrosis post-MI, with concurrent acute increase of heart and chronic increase of splenic neutrophils and monocytes. Mechanistic studies reveal roles of eosinophil IL4 and cationic protein mEar1 in blocking H2O2- and hypoxia-induced mouse and human cardiomyocyte death, TGF-ß-induced cardiac fibroblast Smad2/3 activation, and TNF-α-induced neutrophil adhesion on the heart endothelial cell monolayer. In vitro-cultured eosinophils from WT mice or recombinant mEar1 protein, but not eosinophils from IL4-deficient mice, effectively correct exacerbated cardiac dysfunctions in eosinophil-deficient ∆dblGATA mice. This study establishes a cardioprotective role of eosinophils in post-MI hearts.


Subject(s)
Eosinophils/physiology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/pathology , Aged , Animals , Cell Death , Diphtheria Toxin/toxicity , Electrocardiography , Eosinophils/drug effects , Eosinophils/pathology , Female , Fibroblasts/pathology , Fibroblasts/physiology , Humans , Interleukin-4/genetics , Interleukin-4/metabolism , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/pathology , Ribonucleases/genetics , Ribonucleases/metabolism
12.
FASEB J ; 34(7): 9755-9770, 2020 07.
Article in English | MEDLINE | ID: mdl-32510702

ABSTRACT

Regulatory T cells (Tregs) play essential roles in obesity and diabetes. Here, we report a role of Tregs in enhancing ß3-adrenergic receptor agonist CL316243 (CL)-stimulated thermogenic program in subcutaneous adipose tissue (SAT), but not in visceral fat. CL treatment for 7 days increased SAT adipocyte beiging and thermogenic gene expression in male or female mice. Adoptive transfer of Tregs enhanced this CL activity. Such Treg activity lost in male epididymal white adipose tissue (eWAT) and female gonadal gWAT. Adipocyte culture yielded the same conclusion. Tregs enhanced the expression of CL-induced thermogenic genes in SAT from male and female mice. This activity of Tregs reduced or disappeared in adipocytes from eWAT or gWAT. Both CL and Tregs induced much higher UCP-1 (uncoupling protein-1) expression in SAT from females than that from males. A mechanistic study demonstrated a role of Tregs in suppressing the expression of M1 macrophage markers (Tnfa, Il6, iNos, Ip10) and promoting the expression of M2 macrophage markers (Mrc1, Arg1, Il10) in bone-marrow-derived macrophages or in SAT from male or female mice. In female mice with pre-established obesity, Treg adoptive transfer reduced the gWAT weight in 2 weeks. Together with CL treatment, Treg adoptive transfer reduced the SAT weight and further improved CL-induced glucose metabolism and insulin sensitivity in female obese mice, but did not affect CL-induced body weight loss in male or female obese mice. This study revealed a predominant role of Tregs in female mice in promoting adipocyte beiging and thermogenesis in SAT, in part by slanting M2 macrophage polarization.


Subject(s)
Adipose Tissue, Brown/pathology , Adipose Tissue, White/pathology , Obesity/etiology , Subcutaneous Fat/pathology , T-Lymphocytes, Regulatory/immunology , Thermogenesis , Adipose Tissue, Brown/immunology , Adipose Tissue, White/immunology , Animals , Energy Metabolism , Female , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/pathology , Subcutaneous Fat/immunology , T-Lymphocytes, Regulatory/pathology
13.
Transl Res ; 222: 28-40, 2020 08.
Article in English | MEDLINE | ID: mdl-32434697

ABSTRACT

Nonalcoholic steatohepatitis (NASH) is a severe form of nonalcoholic fatty liver disease characterized by fat accumulation and inflammation in liver. Yet, the mechanistic insight and diagnostic and therapeutic options of NASH remain incompletely understood. This study tested the roles of cysteine protease cathepsin B (CatB) in mouse NASH development. Immunoblot revealed increased liver CatB expression in NASH mice. Fructose-palmitate-cholesterol diet increased body weight gain, liver to body weight ratio, blood fasting glucose, plasma total cholesterol and alanine transaminase levels, and liver triglyceride, but decreased plasma high-density lipoprotein in wild-type mice. All these changes were blunted in CatB-deficient (Ctsb-/-) mice. In parallel to reduced expression of genes involved in liver lipid transport and lipogenesis, liver CD36, FABP4, and PPARγ protein levels were also significantly decreased in Ctsb-/- mice, although CatB deficiency did not affect liver gluconeogenesis and fatty acid beta-oxidation-associated gene expression. Mechanistic studies showed that CatB deficiency decreased liver expression of adhesion molecules, inflammatory cytokine, and chemokine, along with reduced liver inflammatory cell infiltration. CatB deficiency also promoted M2 macrophage polarization and reduced liver TGF-ß1 signaling and fibrosis. Together, CatB deficiency improves liver function in NASH mice by suppressing de novo lipogenesis and liver inflammation and fibrosis.


Subject(s)
Cathepsin B/deficiency , Diet, High-Fat , Inflammation/pathology , Lipid Metabolism , Liver Cirrhosis/complications , Liver Cirrhosis/enzymology , Liver/metabolism , Non-alcoholic Fatty Liver Disease/complications , Animals , Cadherins , Cathepsin B/metabolism , Cell Polarity , Liver/pathology , Liver/physiopathology , Liver Cirrhosis/physiopathology , Macrophages/pathology , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/enzymology , Non-alcoholic Fatty Liver Disease/physiopathology , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Weight Gain
14.
FASEB J ; 34(2): 3091-3104, 2020 02.
Article in English | MEDLINE | ID: mdl-31909541

ABSTRACT

Allergic asthma with high plasma IgE levels is a significant risk factor of human abdominal aortic aneurysm (AAA). This study tests a direct role of IgE in angiotensin-II (Ang-II) perfusion- and peri-aortic CaCl2 injury-induced AAA in mice. In both models, IgE-deficiency in Apoe-/- Ige-/- mice blunts AAA growth and reduces lesion accumulation of macrophages, CD4+ and CD8+ T cells, and lesion MHC class-II expression, CD31+ microvessel growth, and media smooth muscle cell loss, compared with those from Apoe-/- control mice. Real time-PCR reveals significant reductions in expression of neutrophil chemoattractants MIP-2α and CXCL5 in AAA lesions or macrophages from Apoe-/- Ige-/- mice, along with reduced lesion Ly6G+ neutrophil accumulation. Consistent with reduced lesion inflammatory cell accumulation, we find significant reductions of plasma and AAA lesion IL6 expression in Apoe-/- Ige-/- mice. Immunofluorescent staining and FACS analysis show that AAA lesion neutrophils express FcεR1. Mechanistic study demonstrates that IgE induces neutrophil FcεR1 expression, activates MAPK signaling, and promotes IL6 production. This study supports a direct role of IgE in AAA by promoting lesion chemokine expression, inflammatory cell accumulation, MAPK signaling, and cytokine expression. IgE inhibition may represent a novel therapeutic approach in AAA management.


Subject(s)
Aortic Aneurysm, Abdominal/immunology , Immunoglobulin E/deficiency , Neutrophils/metabolism , Animals , Aortic Aneurysm, Abdominal/chemically induced , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/prevention & control , Calcium Chloride/toxicity , Chemokine CXCL2/genetics , Chemokine CXCL2/metabolism , Immunoglobulin E/immunology , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Mice, Knockout, ApoE , Neutrophils/pathology , Receptors, IgE/genetics , Receptors, IgE/metabolism
15.
Radiol Cardiothorac Imaging ; 2(2): e200117, 2020 Apr.
Article in English | MEDLINE | ID: mdl-33778567

ABSTRACT

PURPOSE: To characterize and compare the initial clinical and imaging features of coronavirus disease 2019 (COVID-19) in pediatric and adult patients undergoing chest CT. MATERIALS AND METHODS: A total of 61 patients, consisting of 47 adults (aged 18 years or older) and 14 pediatric patients (aged younger than 18 years) with laboratory-confirmed COVID-19 confirmed by real-time reverse-transcription polymerase chain reaction between January 25 and February 15, 2020, were enrolled in this study. All patients underwent chest CT within 3 days after the initial reverse transcription polymerase chain reaction test. The clinical presentation, serum markers, and CT findings were assessed and compared between the adult and pediatric patients. RESULTS: Fever was less common in pediatric patients than in adults (six of 14, 42.9% vs 39 of 47, 83%; P = .008). Leukopenia or normal, lymphopenia or normal, and increased or normal C-reactive protein level were common in both groups with no difference (P > .05). Compared with the adults, pediatric patients had a lower rate of positive CT findings and a milder clinical grade (P = .004 and P = .001, respectively). At chest CT, the number of pulmonary lobes involved was found to be reduced in pediatric patients when compared with adults (P = .012). Subpleural distribution of lung opacities was a dominant feature in both groups, whereas bronchial distribution was more common in the pediatric group (P = .048). Among the CT features in adults, ground-glass opacities (GGOs) were the most common finding (24 of 43, 53.5%), followed by GGO with consolidation (14 of 43, 27.9%). In pediatric patients, GGOs accounted for 42.9% (three of seven), bronchial wall thickening occurred in 28.6% (two of seven), and GGOs with consolidations and nodular opacities occurred in 14.3% (one of seven). However, these CT features did not differ in the two groups, except for bronchial wall thickening, which was more commonly found in pediatric patients (P = .048). In addition, the semiquantitative scores of lung involvement were higher in adults than in pediatric patients (8.89 ± 4.54 vs 1.86 ± 2.41; P < .001). CONCLUSION: Compared with adults, pediatric patients with COVID-19 showed distinctive clinical and CT features. Pediatric patients tend to have milder clinical symptoms, fewer positive results at CT, and less extensive involvement at imaging. Bronchial wall thickening was relatively more frequent on CT images from pediatric patients with COVID-19 in comparison with adults.Supplemental material is available for this article.© RSNA, 2020.

16.
Transl Res ; 208: 1-14, 2019 06.
Article in English | MEDLINE | ID: mdl-30738862

ABSTRACT

Mast cells (MCs) have been implicated in the pathogenesis of cardiometabolic diseases by releasing pro-inflammatory mediators. Patients and animals with diabetic cardiomyopathy (DCM) also show inflammatory cell accumulation in the heart. Here, we detected MCs in mouse heart after streptozotocin (STZ)-induced DCM. DCM production caused significant systole and diastole interventricular septum and left ventricular (LV) posterior wall thinning, and systolic LV internal dilation in wild-type (WT) mice. DCM production also led to significant reductions of fractional shortening percentage, heart rate, body weight, heart weight, and significant increases of kidney, pancreas, and lung weight to body weight ratios, and blood hemoglobin HbA1c and glucose levels in WT mice. All these changes were improved or disappeared in MC-deficient KitW-sh/W-sh mice. In the myocardium from WT DCM mice, we detected significant decrease of cardiac cell proliferation and increases of cardiac cell death, chemokine expression, macrophage infiltration, inflammatory cytokine expression, and collagen deposition. These changes were also improved or disappeared in KitW-sh/W-sh DCM mice. Adoptive transfer of bone marrow-derived MCs (BMMCs) from WT mice fully or partially reversed these cardiac functional and morphologic changes in KitW-sh/W-sh DCM recipient mice. Yet, adoptive transfer of BMMCs from Il6-/- and Tnf-/- mice failed to make these corrections or at much less extent than the WT BMMCs. Mechanistic studies demonstrated a role of MC and MC-derived IL6 and TNF-α in promoting cardiomyocyte death and cardiac fibroblast TGF-ß signaling, and collagen synthesis and deposition. Therefore, MC inhibition may have therapeutic potential in attenuating DCM progression.


Subject(s)
Diabetic Cardiomyopathies/prevention & control , Mast Cells/pathology , Adoptive Transfer , Animals , Diabetic Cardiomyopathies/pathology , Inflammation/pathology , Interleukin-6/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Streptozocin , Tumor Necrosis Factor-alpha/genetics
17.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1170-1181, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30639224

ABSTRACT

Mouse mast cell protease-4 (mMCP4) is a chymase that has been implicated in cardiovascular diseases, including myocardial infarction (MI). This study tested a direct role of mMCP4 in mouse post-MI cardiac dysfunction and myocardial remodeling. Immunoblot and immunofluorescent double staining demonstrated mMCP4 expression in cardiomyocytes from the infarct zone from mouse heart at 28 day post-MI. At this time point, mMCP4-deficient Mcpt4-/- mice showed no difference in survival from wild-type (WT) control mice, yet demonstrated smaller infarct size, improved cardiac functions, reduced macrophage content but increased T-cell accumulation in the infarct region compared with those of WT littermates. mMCP4-deficiency also reduced cardiomyocyte apoptosis and expression of TGF-ß1, p-Smad2, and p-Smad3 in the infarct region, but did not affect collagen deposition or α-smooth muscle actin expression in the same area. Gelatin gel zymography and immunoblot analysis revealed reduced activities of matrix metalloproteinases and expression of cysteinyl cathepsins in the myocardium, macrophages, and T cells from Mcpt4-/- mice. Immunoblot analysis also found reduced p-Smad2 and p-Smad3 in the myocardium from Mcpt4-/- mice, yet fibroblasts from Mcpt4-/- mice showed comparable levels of p-Smad2 and p-Smad3 to those of WT fibroblasts. Flow cytometry, immunoblot analysis, and immunofluorescent staining demonstrated that mMCP4-deficiency reduced the expression of proapoptotic cathepsins in cardiomyocytes and protected cardiomyocytes from H2O2-induced apoptosis. This study established a role of mMCP4 in mouse post-MI dysfunction by regulating myocardial protease expression and cardiomyocyte death without significant impact on myocardial fibrosis or survival post-MI in mice.


Subject(s)
Myocardial Infarction/metabolism , Myocardium/metabolism , Serine Endopeptidases/deficiency , Ventricular Remodeling , Animals , Apoptosis/genetics , Cells, Cultured , Fibroblasts/metabolism , Fibrosis , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/physiopathology , Myocardium/pathology , Myocytes, Cardiac/metabolism , Serine Endopeptidases/genetics , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
18.
J Mol Cell Cardiol ; 127: 44-56, 2019 02.
Article in English | MEDLINE | ID: mdl-30465799

ABSTRACT

BACKGROUND: Extracellular matrix metabolism and cardiac cell death participate centrally in myocardial infarction (MI). This study tested the roles of collagenolytic cathepsin K (CatK) in post-MI left ventricular remodeling. METHODS AND RESULTS: Patients with acute MI had higher plasma CatK levels (20.49 ±â€¯7.07 pmol/L, n = 26) than those in subjects with stable angina pectoris (8.34 ±â€¯1.66 pmol/L, n = 28, P = .01) or those without coronary heart disease (6.63 ±â€¯0.84 pmol/L, n = 93, P = .01). CatK protein expression increases in mouse hearts at 7 and 28 days post-MI. Immunofluorescent staining localized CatK expression in cardiomyocytes, endothelial cells, fibroblasts, macrophages, and CD4+ T cells in infarcted mouse hearts at 7 days post-MI. To probe the direct participation of CatK in MI, we produced experimental MI in CatK-deficient mice (Ctsk-/-) and their wild-type (Ctsk+/+) littermates. CatK-deficiency yielded worsened cardiac function at 7 and 28 days post-MI, compared to Ctsk+/+ littermates (fractional shortening percentage: 5.01 ±â€¯0.68 vs. 8.62 ±â€¯1.04, P < .01, 7 days post-MI; 4.32 ±â€¯0.52 vs. 7.60 ±â€¯0.82, P < .01, 28 days post-MI). At 7 days post-MI, hearts from Ctsk-/- mice contained less CatK-specific type-I collagen fragments (10.37 ±â€¯1.91 vs. 4.60 ±â€¯0.49 ng/mg tissue extract, P = .003) and more fibrosis (1.67 ±â€¯0.93 vs. 0.69 ±â€¯0.20 type-III collagen positive area percentage, P = .01; 14.25 ±â€¯4.12 vs. 6.59 ±â€¯0.79 α-smooth muscle actin-positive area percentage, P = .016; and 0.82 ±â€¯0.06 vs. 0.31 ±â€¯0.08 CD90-positive area percentage, P = .008) than those of Ctsk+/+ mice. Immunostaining demonstrated that CatK-deficiency yielded elevated cardiac cell death but reduced cardiac cell proliferation. In vitro studies supported a role of CatK in cardiomyocyte survival. CONCLUSION: Plasma CatK levels are increased in MI patients. Heart CatK expression is also elevated post-MI, but CatK-deficiency impairs post-MI cardiac function in mice by increasing myocardial fibrosis and cardiomyocyte death.


Subject(s)
Cathepsin K/deficiency , Heart Function Tests , Myocardial Infarction/enzymology , Myocardial Infarction/physiopathology , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/enzymology , Acute Coronary Syndrome/physiopathology , Aged , Animals , Apoptosis , Cathepsin K/blood , Cell Proliferation , Collagen/metabolism , Female , Fibrosis , Heart Ventricles/metabolism , Humans , Inflammation/pathology , Male , Mice , Middle Aged
19.
Mol Immunol ; 82: 66-74, 2017 02.
Article in English | MEDLINE | ID: mdl-28033540

ABSTRACT

Regulatory T cells (Tregs) are immune suppressive cells, but their roles in tumor growth have been elusive, depending on tumor type or site. Our prior study demonstrated a role of cathepsin S (CatS) in reducing Treg immunosuppressive activity. Therefore, CatS inhibition in Tregs may exacerbate tumor growth. Using mouse bladder carcinoma MB49 cell subcutaneous implant tumor model, we detected no difference in tumor growth, whether mice were given saline- or CatS inhibitor-treated Tregs. However, mice that received inhibitor-treated Tregs had fewer splenic and tumor Tregs, and lower levels of tumor and splenic cell proliferation than mice that received saline-treated Tregs. In vitro, inhibitor-treated Tregs showed lower proliferation and higher apoptosis than saline-treated Tregs when cells were exposed to MB49. In contrast, both types of Tregs showed no difference in proliferation when they were co-cultured with normal splenocytes. Inhibitor-treated Tregs had less apoptosis in splenocytes, but more apoptosis in splenocytes with MB49 conditioned media than saline-treated Tregs. In turn, we detected less proliferation and more apoptosis of MB94 cells after co-culture with inhibitor-treated Tregs, compared with saline-treated Tregs. B220+ B-cell, CD4+ T-cell, and CD8+ T-cell proliferation and apoptosis were also lower in splenocytes co-cultured with inhibitor-treated Tregs than with saline-treated Tregs. Under the same conditions, the addition of cancer cell-conditioned media greatly increased CD8+ T-cell proliferation and reduced CD8+ T-cell apoptosis. These observations suggest that CatS inhibition of Tregs may reduce overall T-cell immunity under normal conditions, but enhance CD8+ T-cell immunity in the presence of cancer cells.


Subject(s)
Carcinoma, Transitional Cell/immunology , Cathepsins/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Urinary Bladder Neoplasms/immunology , Adoptive Transfer , Animals , Apoptosis/immunology , Carcinoma, Transitional Cell/pathology , Cell Proliferation/drug effects , Coculture Techniques , Cysteine Proteases/pharmacology , Disease Models, Animal , Flow Cytometry , Immune Tolerance/immunology , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , T-Lymphocytes, Regulatory/transplantation , Urinary Bladder Neoplasms/pathology
20.
Transl Res ; 171: 1-16, 2016 May.
Article in English | MEDLINE | ID: mdl-26898714

ABSTRACT

Inflammation drives asthma and atherosclerosis. Clinical studies suggest that asthmatic patients have a high risk of atherosclerosis. Yet this hypothesis remains uncertain, given that Th2 imbalance causes asthma whereas Th1 immunity promotes atherosclerosis. In this study, chronic allergic lung inflammation (ALI) was induced in mice by ovalbumin sensitization and challenge. Acute ALI was induced in mice by ovalbumin and aluminum sensitization and ovalbumin challenge. Atherosclerosis was produced in apolipoprotein E-deficient (Apoe(-/-)) mice with a Western diet. When chronic ALI and atherosclerosis were produced simultaneously, ALI increased atherosclerotic lesion size, lesion inflammatory cell content, elastin fragmentation, smooth muscle cell (SMC) loss, lesion cell proliferation, and apoptosis. Production of acute ALI before atherogenesis did not affect lesion size, but increased atherosclerotic lesion CD4(+) T cells, lesion SMC loss, angiogenesis, and apoptosis. Production of acute ALI after atherogenesis also did not change atherosclerotic lesion area, but increased lesion elastin fragmentation, cell proliferation, and apoptosis. In mice with chronic ALI and diet-induced atherosclerosis, daily inhalation of a mast cell inhibitor or corticosteroid significantly reduced atherosclerotic lesion T-cell and mast cell contents, SMC loss, angiogenesis, and cell proliferation and apoptosis, although these drugs did not affect lesion area, compared with those that received vehicle treatment. In conclusion, both chronic and acute ALI promote atherogenesis or aortic lesion pathology, regardless whether ALI occurred before, after, or at the same time as atherogenesis. Antiasthmatic medication can efficiently mitigate atherosclerotic lesion pathology.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/etiology , Hypersensitivity/complications , Pneumonia/complications , Animals , Apolipoproteins E/metabolism , Atherosclerosis/drug therapy , Atherosclerosis/pathology , Budesonide/pharmacology , Budesonide/therapeutic use , Chronic Disease , Disease Progression , Glucocorticoids/pharmacology , Hypersensitivity/drug therapy , Hypersensitivity/pathology , Inflammation/pathology , Ketotifen/pharmacology , Ketotifen/therapeutic use , Male , Mast Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Pneumonia/drug therapy , Pneumonia/pathology
SELECTION OF CITATIONS
SEARCH DETAIL