Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.725
Filter
1.
Cytokine ; 179: 156620, 2024 07.
Article in English | MEDLINE | ID: mdl-38701735

ABSTRACT

PURPOSE: The emergence of immune checkpoint inhibitors (ICIs) has revolutionized cancer treatment, but these drugs can also cause severe immune-related adverse effects (irAEs), including myocarditis. Researchers have become interested in exploring ways to mitigate this side effect, and one promising avenue is the use of baricitinib, a Janus kinase inhibitor known to have anti-inflammatory properties. This study aimed to examine the potential mechanism by which baricitinib in ICIs-related myocarditis. METHODS: To establish an ICIs-related myocarditis model, BALB/c mice were administered murine cardiac troponin I (cTnI) peptide and anti-mouse programmed death 1 (PD-1) antibodies. Subsequently, baricitinib was administered to the mice via intragastric administration. Echocardiography, HE staining, and Masson staining were performed to evaluate myocardial functions, inflammation, and fibrosis. Immunofluorescence was used to detect macrophages in the cardiac tissue of the mice.In vitro experiments utilized raw264.7 cells to induce macrophage polarization using anti-PD-1 antibodies. Different concentrations of baricitinib were applied to assess cell viability, and the release of pro-inflammatory cytokines was measured. The activation of the JAK1/STAT3 signaling pathway was evaluated through western blot analysis. RESULTS: Baricitinib demonstrated its ability to improve cardiac function and reduce cardiac inflammation, as well as fibrosis induced by ICIs. Mechanistically, baricitinib treatment promoted the polarization of macrophages towards the M2 phenotype. In vitro and in vivo experiments showed that anti-PD-1 promoted the release of inflammatory factors. However, treatment with baricitinib significantly inhibited the phosphorylation of JAK1 and STAT3. Additionally, the use of RO8191 reversed the effects of baricitinib, further confirming our findings. CONCLUSION: Baricitinib demonstrated its potential as a protective agent against ICIs-related myocarditis by modulating macrophage polarization. These findings provide a solid theoretical foundation for the development of future treatments for ICIs-related myocarditis.


Subject(s)
Azetidines , Janus Kinase 1 , Macrophages , Mice, Inbred BALB C , Myocarditis , Purines , Pyrazoles , STAT3 Transcription Factor , Sulfonamides , Animals , Male , Mice , Azetidines/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Janus Kinase 1/metabolism , Macrophage Activation/drug effects , Macrophages/metabolism , Macrophages/drug effects , Myocarditis/chemically induced , Myocarditis/drug therapy , Myocarditis/pathology , Myocarditis/metabolism , Purines/pharmacology , Pyrazoles/pharmacology , RAW 264.7 Cells , Signal Transduction/drug effects , STAT3 Transcription Factor/metabolism , Sulfonamides/pharmacology , Troponin I/metabolism
2.
Viruses ; 16(5)2024 04 25.
Article in English | MEDLINE | ID: mdl-38793559

ABSTRACT

Coxsackievirus B3 (CVB3) is a positive single-strand RNA genome virus which belongs to the enterovirus genus in the picornavirus family, like poliovirus. It is one of the most prevalent pathogens that cause myocarditis and pancreatitis in humans. However, a suitable therapeutic medication and vaccination have yet to be discovered. Caboxamycin, a benzoxazole antibiotic isolated from the culture broth of the marine strain Streptomyces sp., SC0774, showed an antiviral effect in CVB3-infected HeLa cells and a CVB3-induced myocarditis mouse model. Caboxamycin substantially decreased CVB3 VP1 production and cleavage of translation factor eIF4G1 from CVB3 infection. Virus-positive and -negative strand RNA was dramatically reduced by caboxamycin treatment. In addition, the cleavage of the pro-apoptotic molecules BAD, BAX, and caspase3 was significantly inhibited by caboxamycin treatment. In animal experiments, the survival rate of mice was improved following caboxamycin treatment. Moreover, caboxamycin treatment significantly decreased myocardial damage and inflammatory cell infiltration. Our study showed that caboxamycin dramatically suppressed cardiac inflammation and mouse death. This result suggests that caboxamycin may be suitable as a potential antiviral drug for CVB3.


Subject(s)
Antiviral Agents , Coxsackievirus Infections , Disease Models, Animal , Enterovirus B, Human , Myocarditis , Animals , Myocarditis/drug therapy , Myocarditis/virology , Mice , Coxsackievirus Infections/drug therapy , Coxsackievirus Infections/virology , Humans , Enterovirus B, Human/drug effects , HeLa Cells , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Male , Mice, Inbred BALB C , Inflammation/drug therapy , Inflammation/virology , Virus Replication/drug effects
3.
Korean J Gastroenterol ; 83(5): 197-199, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38783621

ABSTRACT

5-Aminosalicylic acid (5-ASA) is recommended for managing ulcerative colitis. Common adverse effects associated with 5-ASA include gastrointestinal disorders, headaches, and skin rashes. Perimyocarditis induced by 5-ASA is a rare adverse effect, with only a limited number of cases reported. This paper presents a case of 5-ASA-induced perimyocarditis in a 29-year-old female who had been taking 5-ASA for three weeks. The patient was admitted to the emergency department with dyspnea, chest discomfort, and fever. She subsequently underwent laboratory investigations, including electrocardiography, transthoracic echocardiography, chest computed tomographic angiography, cardiac magnetic resonance imaging, and heart biopsy. Intravenous steroid was administered, and 5-ASA was discontinued. The patient's signs and symptoms improved significantly within a few days of discontinuing 5-ASA, leading to her subsequent discharge. This case highlights the importance of considering perimyocarditis in patients exhibiting cardiac symptoms during 5-ASA therapy, despite it being a rare adverse effect. Drug withdrawal in such cases may lead to rapid clinical improvement.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Colitis, Ulcerative , Echocardiography , Electrocardiography , Mesalamine , Myocarditis , Humans , Female , Mesalamine/therapeutic use , Mesalamine/adverse effects , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/diagnosis , Adult , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Myocarditis/diagnosis , Myocarditis/chemically induced , Myocarditis/drug therapy , Magnetic Resonance Imaging , Tomography, X-Ray Computed , Computed Tomography Angiography
4.
Sci Rep ; 14(1): 9763, 2024 04 29.
Article in English | MEDLINE | ID: mdl-38684719

ABSTRACT

Autoimmune myocarditis is the limited or diffuse inflammation of the myocardium due to dysfunctional cellular and humoral immunity mechanisms. We constructed mouse models of experimental autoimmune myocarditis (EAM) using peptide MyHC-α614-629. On the day after secondary immunization, the mice were intraperitoneally injected with Rho kinase (ROCK) inhibitor Y-27632. On day 21, the cardiac tissues were harvested and weighed. The hearts of EAM mice were significantly enlarged and whitened. Furthermore, body weight (BW) slowly increased during the treatment period, the heart weight (HW) and the ratio of HW/eventual BW were increased, and inflammatory infiltration and fibrosis were aggravated in the myocardial tissue. Y-27632 treatment improved the aforementioned phenotypic and pathological features of EAM mice. Mechanistic analysis revealed a significant increase in Notch1, Hes1, Jag2, Dil1, Toll-like receptor (Tlr) 2, and interleukin (IL)-1ß expression in the myocardial tissue of EAM mice. Notably, IL-1ß expression was correlated with that of Notch1 and Tlr2. Following Y-27632 treatment, the expression of key target genes of the Notch signaling pathway (Notch1, Hes1, Dil1, and Jag2) and Tlr2 were obviously decreased. Y-27632 treatment also decreased the number of monocytes in the spleen of EAM mice. Thus, ROCK inhibitor Y-27632 exerted a protective effect in EAM mice by downregulating IL-1ß expression. This study aimed to provide a reference point for the future treatment of myocarditis in clinical settings.


Subject(s)
Amides , Autoimmune Diseases , Disease Models, Animal , Interleukin-1beta , Myocarditis , Pyridines , rho-Associated Kinases , Animals , Myocarditis/drug therapy , Myocarditis/metabolism , Myocarditis/pathology , Pyridines/pharmacology , Pyridines/therapeutic use , Autoimmune Diseases/drug therapy , Autoimmune Diseases/metabolism , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , Mice , Amides/pharmacology , Amides/therapeutic use , Interleukin-1beta/metabolism , Down-Regulation/drug effects , Male , Myocardium/metabolism , Myocardium/pathology , Signal Transduction/drug effects , Mice, Inbred BALB C
5.
Biomed Pharmacother ; 174: 116535, 2024 May.
Article in English | MEDLINE | ID: mdl-38581923

ABSTRACT

Studies have shown that Sacubitril/valsartan (Sac/Val) can reduce myocardial inflammation in myocarditis mice, in addition to its the recommended treatment of heart failure. However, the underlying mechanisms of Sac/Val in myocarditis remain unclear. C-type natriuretic peptide (CNP), one of the targeting natriuretic peptides of Sac/Val, was recently reported to exert cardio-protective and anti-inflammatory effects in cardiovascular systems. Here, we focused on circulating levels of CNP in patients with acute myocarditis (AMC) and whether Sac/Val modulates inflammation by targeting CNP in experimental autoimmune myocarditis (EAM) mice as well as LPS-induced RAW 264.7 cells and bone marrow derived macrophages (BMDMs) models. Circulating CNP levels were higher in AMC patients compared to healthy controls, and these levels positively correlated with the elevated inflammatory cytokines IL-6 and monocyte count. In EAM mice, Sac/Val alleviated myocardial inflammation while augmenting circulating CNP levels rather than BNP and ANP, accompanied by reduction in intracardial M1 macrophage infiltration and expression of inflammatory cytokines IL-1ß, TNF-α, and IL-6. Furthermore, Sac/Val inhibited CNP degradation and directly blunted M1 macrophage polarization in LPS-induced RAW 264.7 cells and BMDMs. Mechanistically, the effects might be mediated by the NPR-C/cAMP/JNK/c-Jun signaling pathway apart from NPR-B/cGMP/NF-κB pathway. In conclusion, Sac/Val exerts a protective effect in myocarditis by increasing CNP concentration and inhibiting M1 macrophages polarization.


Subject(s)
Aminobutyrates , Biphenyl Compounds , Drug Combinations , Macrophages , Myocarditis , Natriuretic Peptide, C-Type , Valsartan , Animals , Mice , Myocarditis/drug therapy , Myocarditis/metabolism , Myocarditis/pathology , Macrophages/drug effects , Macrophages/metabolism , Aminobutyrates/pharmacology , Valsartan/pharmacology , RAW 264.7 Cells , Male , Humans , Biphenyl Compounds/pharmacology , Natriuretic Peptide, C-Type/pharmacology , Tetrazoles/pharmacology , Acute Disease , Disease Models, Animal , Female , Cytokines/metabolism , Cytokines/blood , Mice, Inbred C57BL , Anti-Inflammatory Agents/pharmacology , Cell Polarity/drug effects
6.
Mol Pharm ; 21(6): 2865-2877, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38666508

ABSTRACT

Imaging strategies for the specific detection and therapeutic monitoring of myocarditis are still lacking. Stimulator of interferon genes (STING) is a signal transduction molecule involved in an innate immune response. Here, we evaluated the feasibility of the recently developed STING-targeted radiotracer [18F]FBTA for positron emission tomography (PET) imaging to detect myocardial inflammation and monitor treatment in myocarditis mice. [18F]FBTA-PET imaging was performed in myocarditis mice and normal mice to verify the specificity of [18F]FBTA for the diagnosis of myocarditis. We also performed PET imaging in mice with myocarditis treated to verify the ability of [18F]FBTA in therapeutic monitoring. The expression of STING and inflammatory cell types was confirmed by flow cytometry and immunohistochemistry. [18F]FDG-PET imaging of myocarditis was used as a contrast. [18F]FBTA-PET imaging showed that the average radioactive uptake was significantly higher in the hearts of the myocarditis group than in the control group. STING was highly overexpressed in cardiac inflammatory cells, including macrophages, dendritic cells (DCs), and T cells. However, there was no significant difference in cardiac radiotracer uptake of [18F]FDG between the myocarditis group and the control group. Moreover, cardiac uptake of [18F]FBTA was significantly reduced in cyclosporin A-treated myocarditis mice and myocardial STING expression was also significantly reduced after the treatment. Overall, we showed that a STING-targeted PET tracer [18F]FBTA can be used to monitor changes in the inflammatory microenvironment in myocarditis. Besides, [18F]FBTA-PET is also suitable for real-time monitoring of myocarditis treatment, representing a promising diagnostic and therapeutic monitoring approach for myocarditis.


Subject(s)
Membrane Proteins , Myocarditis , Positron-Emission Tomography , Myocarditis/diagnostic imaging , Myocarditis/drug therapy , Myocarditis/metabolism , Animals , Mice , Positron-Emission Tomography/methods , Membrane Proteins/metabolism , Male , Radiopharmaceuticals , Fluorodeoxyglucose F18 , Mice, Inbred BALB C , Disease Models, Animal , Myocardium/metabolism , Myocardium/pathology , Dendritic Cells/metabolism , Cyclosporine
7.
ACS Infect Dis ; 10(5): 1793-1807, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38648355

ABSTRACT

Chagas disease, caused by Trypanosoma cruzi, stands as the primary cause of dilated cardiomyopathy in the Americas. Macrophages play a crucial role in the heart's response to infection. Given their functional and phenotypic adaptability, manipulating specific macrophage subsets could be vital in aiding essential cardiovascular functions including tissue repair and defense against infection. PPARα are ligand-dependent transcription factors involved in lipid metabolism and inflammation regulation. However, the role of fenofibrate, a PPARα ligand, in the activation profile of cardiac macrophages as well as its effect on the early inflammatory and fibrotic response in the heart remains unexplored. The present study demonstrates that fenofibrate significantly reduces not only the serum activity of tissue damage biomarker enzymes (LDH and GOT) but also the circulating proportions of pro-inflammatory monocytes (CD11b+ LY6Chigh). Furthermore, both CD11b+ Ly6Clow F4/80high macrophages (MΦ) and recently differentiated CD11b+ Ly6Chigh F4/80high monocyte-derived macrophages (MdMΦ) shift toward a resolving phenotype (CD206high) in the hearts of fenofibrate-treated mice. This shift correlates with a reduction in fibrosis, inflammation, and restoration of ventricular function in the early stages of Chagas disease. These findings encourage the repositioning of fenofibrate as a potential ancillary immunotherapy adjunct to antiparasitic drugs, addressing inflammation to mitigate Chagas disease symptoms.


Subject(s)
Chagas Cardiomyopathy , Fenofibrate , Macrophages , Fenofibrate/pharmacology , Fenofibrate/therapeutic use , Animals , Mice , Chagas Cardiomyopathy/drug therapy , Macrophages/drug effects , Myocardium/pathology , Male , Trypanosoma cruzi/drug effects , Mice, Inbred C57BL , Disease Models, Animal , Myocarditis/drug therapy , Myocarditis/parasitology
8.
Int Immunopharmacol ; 133: 112073, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38636372

ABSTRACT

BACKGROUND: Myocarditis is an important clinical issue which lacks specific treatment by now. Ivermectin (IVM) is an inhibitor of importin α/ß-mediated nuclear translocation. This study aimed to explore the therapeutic effects of IVM on acute myocarditis. METHODS: Mouse models of coxsackie B3 virus (CVB3) infection-induced myocarditis and experimental autoimmune myocarditis (EAM) were established to evaluate the effects of IVM. Cardiac functions were evaluated by echocardiography and Millar catheter. Cardiac inflammatory infiltration was assessed by histological staining. Cytometric bead array and quantitative real-time PCR were used to detect the levels of pro-inflammatory cytokines. The macrophages and their M1/M2 polarization were analyzed via flow cytometry. Protein expression and binding were detected by co-immunoprecipitation, Western blotting and histological staining. The underlying mechanism was verified in vitro using CVB3-infected RAW264.7 macrophages. Cyclic polypeptide (cTN50) was synthesized to selectively inhibit the nuclear translocation of NF-κB/p65, and CVB3-infected RAW264.7 cells were treated with cTN50. RESULTS: Increased expression of importin ß was observed in both models. IVM treatment improved cardiac functions and reduced the cardiac inflammation associated with CVB3-myocarditis and EAM. Furthermore, the pro-inflammatory cytokine (IL-1ß/IL-6/TNF-α) levels were downregulated via the inhibition of the nuclear translocation of NF-κB/p65 in macrophages. IVM and cTN50 treatment also inhibited the nuclear translocation of NF-κB/p65 and downregulated the expression of pro-inflammatory cytokines in RAW264.7 macrophages. CONCLUSIONS: Ivermectin inhibits the nuclear translocation of NF-κB/p65 and the expression of major pro-inflammatory cytokines in myocarditis. The therapeutic effects of IVM on viral and non-viral myocarditis models suggest its potential application in the treatment of acute myocarditis.


Subject(s)
Ivermectin , Mice, Inbred BALB C , Myocarditis , Transcription Factor RelA , Animals , Myocarditis/drug therapy , Myocarditis/virology , Mice , Ivermectin/therapeutic use , Ivermectin/pharmacology , RAW 264.7 Cells , Male , Transcription Factor RelA/metabolism , Coxsackievirus Infections/drug therapy , Enterovirus B, Human/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Cytokines/metabolism , beta Karyopherins/metabolism , Disease Models, Animal , Autoimmune Diseases/drug therapy , Humans , Myocardium/pathology , Myocardium/metabolism
9.
Braz J Infect Dis ; 28(2): 103739, 2024.
Article in English | MEDLINE | ID: mdl-38679059

ABSTRACT

Chlamydia psittaci ‒ related community-acquired pneumonia associated to acute myocarditis was diagnosed in a young man with no medical history, and a professional exposition to birds. The diagnosis was confirmed with positive specific polymerase chain reaction in bronchoalveolar lavage. The patient was treated with spiramycin for two weeks with anti-inflammatory treatment for myocarditis for three months. Clinical and biological improvement was rapidly observed followed by normalization of electrocardiogram and chest CT scan. No relapse was reported for over a two-year follow-up.


Subject(s)
Chlamydophila psittaci , Myocarditis , Psittacosis , Humans , Male , Myocarditis/microbiology , Myocarditis/drug therapy , Myocarditis/diagnostic imaging , Psittacosis/microbiology , Psittacosis/drug therapy , Psittacosis/diagnosis , Chlamydophila psittaci/isolation & purification , Adult , Polymerase Chain Reaction , Community-Acquired Infections/microbiology , Community-Acquired Infections/drug therapy , Acute Disease , Young Adult
11.
J Clin Immunol ; 44(3): 81, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38485795

ABSTRACT

Myocarditis can be caused by viral infection, drug reaction or general inflammatory condition. To provide understanding on inflammatory myocarditis, we describe clinical, genetic, and immunological properties of a young male patient who suffered from recurrent myocarditis episodes since the age of four years. Electrocardiography, troponin I/T, echocardiography, myocardial magnetic resonance imaging and histological findings were consistent with recurrent myocarditis episodes. Homozygous c.245 A > G p.Tyr82Cys pathogenic variant in Hepatitis A Virus Cellular Receptor 2 (HAVCR2) gene encoding T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) receptor was found. Peripheral blood mononuclear cells were collected when the patient was asymptomatic; CD4+ and CD8+ T lymphoblasts, CD56+ natural killer cells and CD14+ monocytes were negative for surface TIM-3 expression. In vitro, TLR4 mediated interleukin-1ß (IL-1ß) response was high after LPS/ATP stimulation. Clinical symptoms responded to IL-1 receptor antagonist anakinra. TIM-3 p.Tyr82Cys CD4+ and CD8+ T cell proliferation in vitro was unrestrained. Findings on IL-2, interferon gamma, regulatory T cells, signal transducer and activator of transcription (STAT) 1, 3 and 4 phosphorylation, and PD-1 and LAG-3 checkpoint inhibitor receptor analyses were comparable to controls. We conclude that TIM-3 deficiency due to homozygous HAVCR2 c.245 A > G p.Tyr82Cys pathogenic variant in the patient described here is associated with autoinflammatory symptoms limited to early onset recurrent febrile myocarditis. Excessive IL-1ß production and defective regulation of T cell proliferation may contribute to this clinical condition responsive to anakinra treatment.


Subject(s)
Hepatitis A Virus Cellular Receptor 2 , Myocarditis , Humans , Male , Child, Preschool , Hepatitis A Virus Cellular Receptor 2/genetics , Myocarditis/diagnosis , Myocarditis/drug therapy , Myocarditis/etiology , Leukocytes, Mononuclear , Interleukin 1 Receptor Antagonist Protein , Interleukin-1beta , Germ Cells
12.
Phytomedicine ; 128: 155558, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38547614

ABSTRACT

BACKGROUND: The experimental autoimmune myocarditis (EAM) model is valuable for investigating myocarditis pathogenesis. M1-type macrophages and CD4+T cells exert key pathogenic effects on EAM initiation and progression. Baicalein (5,6,7-trihydroxyflavone, C15H10O5, BAI), which is derived from the Scutellaria baicalensis root, is a primary bioactive compound with potent anti-inflammatory and antioxidant properties. BAI exerts good therapeutic effects against various autoimmune diseases; however, its effect in EAM has not been thoroughly researched. PURPOSE: This study aimed to explore the possible inhibitory effect of BAI on M1 macrophage polarisation and CD4+T cell differentiation into Th1 cells via modulation of the JAK-STAT1/4 signalling pathway, which reduces the secretion of pro-inflammatory factors, namely, TNF-α and IFN-γ, and consequently inhibits TNF-α- and IFN-γ-triggered apoptosis in cardiomyocytes of the EAM model mice. STUDY DESIGN AND METHODS: Flow cytometry, immunofluorescence, real-time quantitative polymerase chain reaction (q-PCR), and western blotting were performed to determine whether BAI alleviated M1/Th1-secreted TNF-α- and IFN-γ-induced myocyte death in the EAM model mice through the inhibition of the JAK-STAT1/4 signalling pathway. RESULTS: These results indicate that BAI intervention in mice resulted in mild inflammatory infiltrates. BAI inhibited JAK-STAT1 signalling in macrophages both in vivo and in vitro, which attenuated macrophage polarisation to the M1 type and reduced TNF-α secretion. Additionally, BAI significantly inhibited the differentiation of CD4+T cells to Th1 cells and IFN-γ secretion both in vivo and in vitro by modulating the JAK-STAT1/4 signalling pathway. This ultimately led to decreased TNF-α and IFN-γ levels in cardiac tissues and reduced myocardial cell apoptosis. CONCLUSION: This study demonstrates that BAI alleviates M1/Th1-secreted TNF-α- and IFN-γ-induced cardiomyocyte death in EAM mice by inhibiting the JAK-STAT1/4 signalling pathway.


Subject(s)
Apoptosis , Disease Models, Animal , Flavanones , Interferon-gamma , Janus Kinases , Myocarditis , Myocytes, Cardiac , STAT1 Transcription Factor , Signal Transduction , Tumor Necrosis Factor-alpha , Animals , STAT1 Transcription Factor/metabolism , Signal Transduction/drug effects , Myocytes, Cardiac/drug effects , Janus Kinases/metabolism , Mice , Flavanones/pharmacology , Male , Interferon-gamma/metabolism , Apoptosis/drug effects , Tumor Necrosis Factor-alpha/metabolism , Myocarditis/drug therapy , STAT4 Transcription Factor/metabolism , Autoimmune Diseases/drug therapy , Mice, Inbred BALB C , Macrophages/drug effects , Macrophages/metabolism , Scutellaria baicalensis/chemistry , Th1 Cells/drug effects , Cell Differentiation/drug effects
13.
Pediatr Cardiol ; 45(5): 1048-1054, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38509207

ABSTRACT

The purpose of this paper was to unravel the clinical effect analysis of different doses of creatine phosphate sodium (CPS) combined with immunoglobulin in the treatment of pediatric viral myocarditis (VMC). One hundred and twenty children with VMC were recruited and randomized into three groups (40 patients each). Group I received 1.0 g of CPS dissolved in 100 mL of 5% glucose injection intravenously 1 time/day; group II received 1.25 g of CPS dissolved in 125 mL of 5% glucose injection intravenously 1 time/day; group III received 1.5 g of CPS dissolved in 150 mL of 5% glucose injection intravenously 1 time/day; then all three groups were treated with combined use of immunoglobulin (300-400 mg/day) intravenously once a day; and all three groups were treated for 14 days. The clinical efficacy, cardiac function, serum inflammatory factor levels, immune function, and the occurrence of drug toxicity and adverse effects of the children in the three groups were compared after 14 days of treatment. All three groups achieved better therapeutic effects after treatment, in which the effective rate of the Group II and Group III was notably higher versus the Group I. Lower levels of cTnI, CK-MB, LDH, AST, IL-18, IL-6, IFN-γ, and LVEDD and higher CD3+, CD4+, and CD4+/CD8+, FS, and LVEF values were noted in the Group II and Group III versus the Group I, and the results were more pronounced in the high-dose group. The liver and kidney functions of the children in the three groups before and after treatment did not show any significant changes and the incidence of adverse reactions during the treatment period was low in all three groups. Children with VMC can be treated with high-dose CPS in combination with immunoglobulin, which can improve their cardiac function and immune function and reduce the inflammatory response with good overall therapeutic efficacy and fewer adverse effects.


Subject(s)
Myocarditis , Phosphocreatine , Humans , Myocarditis/drug therapy , Male , Female , Child , Child, Preschool , Treatment Outcome , Drug Therapy, Combination , Dose-Response Relationship, Drug , Virus Diseases/drug therapy , Immunoglobulins/administration & dosage , Immunoglobulins/therapeutic use
14.
ESMO Open ; 9(2): 102383, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38364453

ABSTRACT

BACKGROUND: Treatment with tumor-infiltrating lymphocytes (TILs) is rapidly evolving for patients with solid tumors. Following metastasectomy, TILs (autologous, intratumoral CD4+ and CD8+ T cells with the potential to recognize tumor-associated antigens) are isolated and non-specifically expanded ex vivo in the presence of interleukin-2 (IL-2). Subsequently, the TILs are adoptively transferred to the patients after a preconditioning non-myeloablative, lymphodepleting chemotherapy regimen, followed by administration of high-dose (HD) IL-2. Here, we provide an overview of known cardiac risks associated with TIL treatment and report on seven patients presenting with cardiac symptoms, all with different clinical course and diagnostic findings during treatment with lymphodepleting chemotherapy, TIL, and HD IL-2, and propose a set of clinical recommendations for diagnosis and management of these symptoms. PATIENTS AND METHODS: This single-center, retrospective study included selected patients who experienced TIL treatment-related cardiac symptoms at the Netherlands Cancer Institute. In addition, 12 patients were included who received TIL in the clinical trial setting without experiencing cardiac symptoms, from whom complete cardiac biomarker follow-up during treatment was available [creatine kinase (CK), CK-myocardial band, troponin T and N-terminal pro-B-type natriuretic peptide]. RESULTS: Within our TIL patient population, seven illustrative cases were chosen from the patients who developed symptoms suspected of severe cardiotoxicity: myocarditis, myocardial infarction, peri-myocarditis, atrial fibrillation, acute dyspnea, and two cases of heart failure. An overview of their clinical course, diagnostics carried out, and management of the symptoms is provided. CONCLUSIONS: In the absence of evidence-based guidelines for the treatment of TIL therapy-associated cardiotoxicity, we provided an overview of literature, case descriptions, and recommendations for diagnosis and management to help physicians in daily practice, as the number of patients qualifying for TIL treatment is rapidly increasing.


Subject(s)
Lymphocytes, Tumor-Infiltrating , Myocarditis , Humans , Lymphocytes, Tumor-Infiltrating/pathology , Interleukin-2/therapeutic use , Myocarditis/drug therapy , Myocarditis/pathology , Retrospective Studies , Disease Progression
17.
Mol Immunol ; 167: 43-52, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38354482

ABSTRACT

OBJECTIVE: To investigate the anti-inflammatory actions and molecular mechanisms of the sodium/glucose cotransporter 2 (SGLT-2) inhibitor empagliflozin on autoimmune myocarditis. METHODS: The experimental autoimmune myocarditis (EAM) mouse model was constructed using peptides, and the therapeutic effects of empagliflozin on cardiac inflammation and fibrosis were observed using hematoxylin and eosin (HE), Sirius red staining, and Masson's trichome staining. Western blotting was used to identify the actions of empagliflozin on the surface marker expression levels of M2 macrophages and inflammatory factors. In vitro, experiments were completed using lentiviral overexpression of SGLT-2 in macrophages. Macrophage inflammation and anti-inflammatory models were constructed using lipopolysaccharide and interleukin-4, respectively. Enzyme-linked immunosorbent assay, immunofluorescence staining, and reverse-transcription polymerase chain reaction were applied to detect the effects of empagliflozin on the levels of inflammatory factors and macrophage surface markers. Western blotting was used to identify variability in SGLT-2 expression and the role of empagliflozin on the signal transducer and activator of the transcription 3 (STAT3) pathway. The Genomic Spatial Event 142564 dataset was studied in an EAM mouse model. We selected single-cell sequencing results from day 0 and day 21 of modeling to visualize differentially expressed genes. Immune cell infiltration correlation analysis was implemented to explore the expression of inflammatory factors and phenotypic markers. RESULTS: Empagliflozin increased the expression of the M2 macrophage surface marker CD206 and reduced the level of inflammatory factors in the EAM mouse model while reducing the levels of inflammation and fibrosis. In vitro experiments revealed that the phosphorylation of STAT3 pathway was enhanced after macrophages were polarized to M1 phenotype by LPS, the phosphorylation of STAT3 pathway was inhibited after empagliflozin intervention, and the levels of inflammatory factors were decreased. CONCLUSION: Empagliflozin can reduce the level of inflammation in autoimmune myocarditis through the STAT3 pathway and macrophage phenotype transformation. These results indicate the expression of SGLT-2 can be a target for autoimmune myocarditis therapy.


Subject(s)
Benzhydryl Compounds , Glucosides , Myocarditis , Mice , Animals , Myocarditis/drug therapy , Inflammation , Macrophages/metabolism , Phenotype , Anti-Inflammatory Agents/pharmacology , Fibrosis
18.
J Immunother Cancer ; 12(1)2024 01 17.
Article in English | MEDLINE | ID: mdl-38233099

ABSTRACT

Immune checkpoint inhibitor (ICI) treatment has become an important therapeutic option for various cancer types. Although the treatment is effective, ICI can overstimulate the patient's immune system, leading to potentially severe immune-related adverse events (irAEs), including hepatitis, colitis, pneumonitis and myocarditis. The initial mainstay of treatments includes the administration of corticosteroids. There is little evidence how to treat steroid-resistant (sr) irAEs. It is mainly based on small case series or single case reports. This systematic review summarizes available evidence about sr-irAEs. We conducted a systematic literature search in PubMed. Additionally, we included European Society for Medical Oncology, Society for Immunotherapy of Cancer, National Comprehensive Cancer Network and American Society of Clinical Oncology Guidelines for irAEs in our assessment. The study population of all selected publications had to include patients with cancer who developed hepatitis, colitis, pneumonitis or myocarditis during or after an immunotherapy treatment and for whom corticosteroid therapy was not sufficient. Our literature search was not restricted to any specific cancer diagnosis. Case reports were also included. There is limited data regarding life-threatening sr-irAEs of colon/liver/lung/heart and the majority of publications are single case reports. Most publications investigated sr colitis (n=26), followed by hepatitis (n=21), pneumonitis (n=17) and myocarditis (n=15). There is most data for mycophenolate mofetil (MMF) to treat sr hepatitis and for infliximab, followed by vedolizumab, to treat sr colitis. Regarding sr pneumonitis there is most data for MMF and intravenous immunoglobulins (IVIG) while data regarding infliximab are conflicting. In sr myocarditis, most evidence is available for the use of abatacept or anti-thymocyte globulin (ATG) (both with or without MMF) or ruxolitinib with abatacept. This review highlights the need for prompt recognition and treatment of sr hepatitis, colitis, pneumonitis and myocarditis. Guideline recommendations for sr situations are not defined precisely. Based on our search, we recommend-as first line treatment-(1) MMF for sr hepatitis, (2) infliximab for sr colitis, followed by vedolizumab, (3) MMF and IVIG for sr pneumonitis and (4) abatacept or ATG (both with or without MMF) or ruxolitinib with abatacept for sr myocarditis. These additional immunosuppressive agents should be initiated promptly if there is no sufficient response to corticosteroids within 3 days.


Subject(s)
Colitis , Hepatitis , Myocarditis , Neoplasms , Nitriles , Pneumonia , Pyrazoles , Pyrimidines , Humans , Abatacept/therapeutic use , Adrenal Cortex Hormones/therapeutic use , Colitis/chemically induced , Colitis/drug therapy , Hepatitis/drug therapy , Immunoglobulins, Intravenous/therapeutic use , Infliximab/therapeutic use , Mycophenolic Acid/therapeutic use , Myocarditis/drug therapy , Neoplasms/complications , Neoplasms/drug therapy , Pneumonia/drug therapy
20.
ACS Nano ; 18(4): 2782-2799, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38232382

ABSTRACT

Immune regulation therapies are considered promising for treating classically activated macrophage (M1)-driven viral myocarditis (VM). Alternatively, activated macrophage (M2)-derived extracellular vesicles (M2 EVs) have great immunomodulatory potential owing to their ability to reprogram macrophages, but their therapeutic efficacy is hampered by insufficient targeting capacity in vivo. Therefore, we developed cardiac-targeting peptide (CTP) and platelet membrane (PM)-engineered M2 EVs enriched with viral macrophage inflammatory protein-II (vMIP-II), termed CTP/PM-M2 EVsvMIP-II-Lamp2b, to improve the delivery of EVs "cargo" to the heart tissues. In a mouse model of VM, the intravenously injected CTP/PM-M2 EVsvMIP-II-Lamp2b could be carried into the myocardium via CTP, PM, and vMIP-II. In the inflammatory microenvironment, macrophages differentiated from circulating monocytes and macrophages residing in the heart showed enhanced endocytosis rates for CTP/PM-M2 EVsvMIP-II-Lamp2b. Subsequently, CTP/PM-M2 EVsvMIP-II-Lamp2b successfully released functional M2 EVsvMIP-II-Lamp2b into the cytosol, which facilitated the reprogramming of inflammatory M1 macrophages to reparative M2 macrophages. vMIP-II not only helps to increase the targeting ability of M2 EVs but also collaborates with M2 EVs to regulate M1 macrophages in the inflammatory microenvironment and downregulate the levels of multiple chemokine receptors. Finally, the cardiac immune microenvironment was protectively regulated to achieve cardiac repair. Taken together, our findings suggest that CTP-and-PM-engineered M2 EVsvMIP-II-Lamp2b represent an effective means for treating VM and show promise for clinical applications.


Subject(s)
Extracellular Vesicles , Myocarditis , Mice , Animals , Myocarditis/drug therapy , Macrophages , Monocytes , Phagocytosis
SELECTION OF CITATIONS
SEARCH DETAIL
...