Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Circulation ; 150(8): 622-641, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38660786

ABSTRACT

BACKGROUND: Dysregulated metabolism of bioactive sphingolipids, including ceramides and sphingosine-1-phosphate, has been implicated in cardiovascular disease, although the specific species, disease contexts, and cellular roles are not completely understood. Sphingolipids are produced by the serine palmitoyltransferase enzyme, canonically composed of 2 subunits, SPTLC1 (serine palmitoyltransferase long chain base subunit 1) and SPTLC2 (serine palmitoyltransferase long chain base subunit 2). Noncanonical sphingolipids are produced by a more recently described subunit, SPTLC3 (serine palmitoyltransferase long chain base subunit 3). METHODS: The noncanonical (d16) and canonical (d18) sphingolipidome profiles in cardiac tissues of patients with end-stage ischemic cardiomyopathy and in mice with ischemic cardiomyopathy were analyzed by targeted lipidomics. Regulation of SPTLC3 by HIF1α under ischemic conditions was determined with chromatin immunoprecipitation. Transcriptomics, lipidomics, metabolomics, echocardiography, mitochondrial electron transport chain, mitochondrial membrane fluidity, and mitochondrial membrane potential were assessed in the cSPTLC3KO transgenic mice we generated. Furthermore, morphological and functional studies were performed on cSPTLC3KO mice subjected to permanent nonreperfused myocardial infarction. RESULTS: Herein, we report that SPTLC3 is induced in both human and mouse models of ischemic cardiomyopathy and leads to production of atypical sphingolipids bearing 16-carbon sphingoid bases, resulting in broad changes in cell sphingolipid composition. This induction is in part attributable to transcriptional regulation by HIF1α under ischemic conditions. Furthermore, cardiomyocyte-specific depletion of SPTLC3 in mice attenuates oxidative stress, fibrosis, and hypertrophy in chronic ischemia, and mice demonstrate improved cardiac function and increased survival along with increased ketone and glucose substrate metabolism utilization. Depletion of SPTLC3 mechanistically alters the membrane environment and subunit composition of mitochondrial complex I of the electron transport chain, decreasing its activity. CONCLUSIONS: Our findings suggest a novel essential role for SPTLC3 in electron transport chain function and a contribution to ischemic injury by regulating complex I activity.


Subject(s)
Cardiomyopathies , Electron Transport Complex I , Serine C-Palmitoyltransferase , Animals , Serine C-Palmitoyltransferase/metabolism , Serine C-Palmitoyltransferase/genetics , Mice , Humans , Cardiomyopathies/metabolism , Cardiomyopathies/genetics , Electron Transport Complex I/metabolism , Electron Transport Complex I/genetics , Sphingolipids/metabolism , Mice, Knockout , Myocardial Ischemia/metabolism , Myocardial Ischemia/genetics , Myocardial Ischemia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology
2.
Methods Mol Biol ; 2641: 101-113, 2023.
Article in English | MEDLINE | ID: mdl-37074644

ABSTRACT

The NACHT, LRR, and PYD domain-containing protein-3 (NLRP3) inflammasome activation is part of a stereotyped cellular response to injury or infection. The NLRP3 inflammasome activation promotes cellular dysfunction and death, leading to local and systemic inflammation, organ dysfunction, and adverse outcome. Immunohistochemistry and immunofluorescence can be used to determine whether the NLRP3 inflammasome components are present in human biopsy or autopsy tissue samples.


Subject(s)
Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Humans , Inflammation , Interleukin-1beta
3.
Transl Res ; 252: 9-20, 2023 02.
Article in English | MEDLINE | ID: mdl-35948198

ABSTRACT

Despite significant advances and the continuous development of novel, effective therapies to treat a variety of malignancies, cancer therapy-induced cardiotoxicity has been identified as a prominent cause of morbidity and mortality, closely competing with secondary malignancies. This unfortunate limitation has prompted the inception of the field of cardio-oncology with its purpose to provide the necessary knowledge and key information on mechanisms that support the use of the most efficacious cancer therapy with minimal or no interruption while paying close attention to preventing cardiovascular related morbidity and mortality. Several mechanisms that contribute to cancer therapy-induced cardiotoxicity have been proposed and studied. These mainly involve mitochondrial dysfunction and reactive oxygen species-induced oxidative stress, lysosomal damage, impaired autophagy, cell senescence, DNA damage, and sterile inflammation with the formation and activation of the NLRP3 inflammasome. In this review, we focus on describing the principal mechanisms for different classes of cancer therapies that lead to cardiotoxicity involving the NLRP3 inflammasome. We also summarize current evidence of cardio-protection with inflammasome inhibitors in the context of heart disease in general, and further highlight the potential application of this evidence for clinical translation in at risk patients for the purpose of preventing cancer therapy associated cardiovascular morbidity and mortality.


Subject(s)
Inflammasomes , Neoplasms , Humans , NLR Family, Pyrin Domain-Containing 3 Protein , Cardiotoxicity/etiology , Inflammation , Neoplasms/complications , Neoplasms/drug therapy
4.
Rev Cardiovasc Med ; 24(3): 77, 2023 Mar.
Article in English | MEDLINE | ID: mdl-39077487

ABSTRACT

Acute pericarditis is the most frequent pericardial disease characterized by inflammation of the pericardial layers resulting in pain, dyspnea and fatigue. Often limited to an isolated event, up to 30% of patients experience one or more recurrences. There is limited knowledge about the pathophysiology of this disease, possibly due to the limited availability of animal models. More recently, following seminal clinical trials with colchicine and interleukin-1 (IL-1) blockers and a novel murine model of acute pericarditis using zymosan A, it has become clear that the NLRP3 (NACHT, leucine-rich repeat, and pyrin domain-containing protein 3) inflammasome/IL-1 ß axis plays a central role in driving acute pericardial inflammation and in sustaining this process during recurrences. Diagnostic management of pericarditis has been implemented with multimodality imaging including echocardiography, cardiac computed tomography, and cardiac magnetic resonance. These imaging modalities provide essential diagnostic and pathogenetic information, and are able to characterize pericardial inflammation, allowing to refine risk stratification and personalize treatment. Recent acquisitions yield relevant implications with regard to the therapeutic management of acute and recurrent pericarditis. Non-steroidal anti-inflammatory drugs (NSAIDs) and colchicine are cornerstone therapies either for acute and recurrent pericarditis. However, the benefits of targeted agents, such as anakinra - a recombinant human IL-1 receptor antagonist - and rilonacept - an IL-1 α /IL-1 ß trap, are being increasingly recognized. To this end, phenotyping patients with pericarditis and addressing such therapies to those presenting with auto-inflammatory features (elevated C-reactive protein, sustained pericardial and systemic inflammation, multiple recurrences) is of utmost importance to identify patients who might be more likely to benefit from NLRP3 inflammasome/IL-1 ß pathway blockade.

5.
JACC CardioOncol ; 5(6): 715-731, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38205010

ABSTRACT

Despite improvements in cancer survival, cancer therapy-related cardiovascular toxicity has risen to become a prominent clinical challenge. This has led to the growth of the burgeoning field of cardio-oncology, which aims to advance the cardiovascular health of cancer patients and survivors, through actionable and translatable science. In these Global Cardio-Oncology Symposium 2023 scientific symposium proceedings, we present a focused review on the mechanisms that contribute to common cardiovascular toxicities discussed at this meeting, the ongoing international collaborative efforts to improve patient outcomes, and the bidirectional challenges of translating basic research to clinical care. We acknowledge that there are many additional therapies that are of significance but were not topics of discussion at this symposium. We hope that through this symposium-based review we can highlight the knowledge gaps and clinical priorities to inform the design of future studies that aim to prevent and mitigate cardiovascular disease in cancer patients and survivors.

6.
JACC CardioOncol ; 4(4): 549-551, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36444230
7.
Adv Cancer Res ; 155: 167-214, 2022.
Article in English | MEDLINE | ID: mdl-35779874

ABSTRACT

The quest of defeating cancer and improving prognosis in survivors has generated remarkable strides forward in research and have advanced the development of new antineoplastic therapies. These achievements, combined with rapid screening and early detection, have considerably extended the life expectancy of patients surviving multiple types of malignancies. Consequently, chemotherapy-related toxicity in several organ systems, especially the cardiovascular system, has surfaced as one of the leading causes of morbidity and mortality among cancer survivors. Recent evidence classifies chemotherapy-induced cardiotoxicity as the second-leading cause of morbidity and mortality, closely comparing with secondary cancer malignancies. While a certain degree of cardiotoxicity has been reported to accompany most chemotherapies, including anthracyclines, anti-metabolites, and alkylating agents, even the latest targeted cancer therapies such as immune checkpoint inhibitors and tyrosine kinase inhibitors have been associated with acute and chronic cardiac sequelae. In this chapter, we focus on describing the principal mechanism(s) for each class of chemotherapeutic agents that lead to cardiotoxicity and the innovative translational research approaches that are currently being explored to prevent or treat cancer therapy-induced cardiotoxicity and related cardiac complications.


Subject(s)
Cancer Survivors , Neoplasms , Cardiotoxicity/etiology , Humans , Neoplasms/drug therapy , Translational Research, Biomedical
8.
Sci Rep ; 12(1): 8897, 2022 05 25.
Article in English | MEDLINE | ID: mdl-35614179

ABSTRACT

Relaxin is a pleiotropic hormone demonstrated to confer cardioprotection in animal models of myocardial infarction and ischemic heart failure by modulating inflammation, fibrosis and arrhythmogenesis. Several of these pathways in the ischemic myocardium are intricately tied with the downstream signaling of bioactive sphingolipids, which play an active role during post-infarction remodeling. In this current study, we examined the effects of relaxin on sphingosine 1-phosphate (S1P), and the potential benefits of relaxin treatment on cardiac health in a rodent model of ischemic heart failure. Acute (30 min) and sub-acute (24 h) treatment of primary cardiomyocytes with serelaxin (recombinant human relaxin-2) increased the cardiomyocyte content of S1P. In the rodent model, treatment with relaxin for 28 days following myocardial ischemia by way of permanent left coronary artery occlusion improved survival and cardiac function, reduced fibrosis and apoptosis, and mitigated the expression of several pro-inflammatory and pro-fibrotic markers. The expression of beclin-1 (autophagy marker) was also reduced. The expression of S1P was significantly higher in cardiac tissue and plasma samples extracted from serelaxin-treated mice at day 28. In conclusion, our studies show a significant protection from relaxin in ischemic heart disease, and demonstrate the association between relaxin signaling and S1P generation.


Subject(s)
Heart Failure , Myocardial Ischemia , Relaxin , Animals , Disease Models, Animal , Fibrosis , Heart Failure/drug therapy , Heart Failure/metabolism , Mice , Myocardial Ischemia/drug therapy , Myocardial Ischemia/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Relaxin/metabolism , Relaxin/pharmacology , Signal Transduction , Sphingolipids/metabolism
9.
Expert Opin Pharmacother ; 23(6): 681-691, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35311434

ABSTRACT

INTRODUCTION: Aspirin or non-steroidal anti-inflammatory drugs (NSAIDs) and colchicine are first-line treatments for acute and recurrent pericarditis. Drugs blocking the NACHT, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome/interleukin-1ß (IL-1ß) axis are beneficial in patients with multiple recurrences. AREAS COVERED: In this review, the role of the NLRP3 inflammasome/IL-1ß axis in the pathophysiology of pericarditis is discussed. Updates about novel therapies targeting IL-1 for recurrent pericarditis (RP) and practical considerations for their use are provided. EXPERT OPINION: IL-1 inhibitors have been increasingly studied for RP in recent years. NLRP3 inflammasome is a key mediator in the pathophysiology of RP. IL-1ß, its main product, can sustain its own production and feeds local and systemic inflammation. Randomized clinical trials testing anakinra (a recombinant form of the IL-1 receptor antagonist blocking IL-1α and IL-1ß) and rilonacept (an IL-1α and IL-1ß trap) have shown that IL-1 blockade reduces recurrences. These trials also helped in phenotyping patients with RP. Patients with multiple recurrences and signs of pericardial and/or systemic inflammation might benefit from IL-1 blockers in order to interrupt cyclic flares of auto-inflammation. Given this evidence, guidelines should consider incorporating IL-1 blockers.


Subject(s)
Pericarditis , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Colchicine/therapeutic use , Humans , Inflammasomes/metabolism , Inflammasomes/therapeutic use , Inflammation/drug therapy , Pericarditis/diagnosis , Pericarditis/drug therapy
10.
Artif Organs ; 46(7): 1346-1357, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35167122

ABSTRACT

BACKGROUND: Donation after circulatory death (DCD) hearts requires machine perfusion preservation, the conditions of which are not well defined. METHODS: To achieve this, rat hearts were procured following a DCD or control beating-heart donation (CBD) model, and perfused for 60 min with one of three machine perfusion solutions-St. Thomas (ST), University of Wisconsin (UW), or Polyethylene Glycol-20k (PEG)-at one of two temperatures, 4°C or 15°C. At 15-min intervals, perfusion pressure was measured as a marker of vascular resistance. Colored microspheres were added to capture the distribution of perfusate into the metabolically active sub-endocardium, and the eluate was collected for troponin assays. Analyses compared groups using Wilcoxon rank-sum and ANOVA. RESULTS: Perfusion pressure was significantly higher for DCD than CBD hearts at 15°C regardless of solutions. The lowest rise in perfusion pressure over time was observed with PEG at 15°C. Except for PEG at 15°C, ST and UW solutions at 4 or 15°C had decreased sub-endocardial perfusion in DCD hearts. Troponin release from DCD hearts with UW and PEG solutions was comparable to CBD hearts but was significantly higher with ST solution at 15°C. CONCLUSIONS: Optimal preservation conditions for DCD hearts were observed with PEG machine perfusion solution at 15°C.


Subject(s)
Heart Transplantation , Organ Preservation Solutions , Animals , Heart , Humans , Organ Preservation , Organ Preservation Solutions/pharmacology , Perfusion , Rats , Troponin
11.
JACC Basic Transl Sci ; 7(1): 53-63, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35128209

ABSTRACT

Relaxin is a pleiotropic hormone shown to confer cardioprotection in several preclinical models of cardiac ischemia-reperfusion injury. In the present study, the effects of up-regulating relaxin family peptide receptor 1 (RXFP1) via adeno-associated virus serotype 9 (AAV9) vectors were investigated in a mouse model of myocardial infarction. AAV9-RXFP1 vectors were generated and injected in adult male CD1 mice. Up-regulation of Rxfp1 was confirmed via quantitative polymerase chain reaction, and overexpressing animals showed increased sensitivity to relaxin-induced ventricular inotropic response. Overexpressing animals also demonstrated reduced infarct size and preserved cardiac function 24 hours after ischemia-reperfusion. Up-regulation of RXFP1 via AAV9 vectors has potential therapeutic utility in preventing adverse remodeling after myocardial infarction.

12.
Cytokine ; 151: 155811, 2022 03.
Article in English | MEDLINE | ID: mdl-35091314

ABSTRACT

BACKGROUND: Aging is associated with metabolic and structural changes causing heart failure with preserved ejection fraction (HFpEF). Interleukin-1 (IL-1) is a pro-inflammatory cytokine involved in aging-related inflammation. OBJECTIVE: We sought to determine whether IL-1 mediates aging-related changes in the heart, as seen in HFpEF. METHODS: We studied age-matched young (4-month-old), middle-aged (14-month-old), and old (23-month-old) wild-type (WT) C57BL/6J and IL-1 receptor type I deficient (IL1RI-KO) male mice. Echocardiography was used to evaluate left ventricular (LV) dimensions and systolic/diastolic function, and a pressure transducer was used to measure the LV end-diastolic pressure. Picrosirius red stain was used to assess for myocardial interstitial fibrosis (MIF) at pathology. RESULTS: WT and IL-1RIKO mice showed a normal cardiac phenotype at young age, without any differences between the two groups. With aging, the WT mice developed LV concentric hypertrophy (as measured by a significant increase in LV mass [+42%, P < 0.01] and relative wall thickness [+34%, P < 0.01]), whereas the aging IL-1RI-KO mice did not. With aging, the WT mice also developed diastolic dysfunction (as measured by a significant increase in isovolumetric relaxation time [+148%, P < 0.01] and a significantly higher LV end-diastolic pressure [+174%, P < 0.01]), whereas the aging IL1RI-KO did not. Aged WT mice showed a significant increase in MIF (+124%, P < 0.01) at cardiac pathology, whereas the aging IL-1RI-KO did not. CONCLUSIONS: Genetically-modified mice lacking the IL-1RI receptor, not responsive to IL-1, are protected from aging-related LV hypertrophy, fibrosis, and diastolic dysfunction. These data support a central role of IL-1 in the pathophysiology of aging-related HFpEF.


Subject(s)
Cardiomyopathies , Heart Failure , Receptors, Interleukin-1 Type I , Age Factors , Aging , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , Male , Mice , Mice, Inbred C57BL , Receptors, Interleukin-1 Type I/metabolism , Stroke Volume/physiology , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology
13.
Animal Model Exp Med ; 4(3): 283-296, 2021 09.
Article in English | MEDLINE | ID: mdl-34557655

ABSTRACT

Heart transplantation is a lifesaving procedure, which is limited by the availability of donor hearts. Using hearts from donors after circulatory death, which have sustained global ischemia, requires thorough studies on reliable and reproducible models that developing researchers may not have mastered. By combining the most recent literature and our recommendations based on observations and trials and errors, the methods here detail a sound in vivo heterotopic heart transplantation model for rats in which protective interventions on the ischemic heart can be studied, and thus allowing the scientific community to advance organ preservation research. Knowledge gathered from reproducible animal models allow for successful translation to clinical studies.


Subject(s)
Heart Transplantation , Reperfusion Injury , Animals , Heart Transplantation/methods , Humans , Ischemia , Mice , Rats , Tissue Donors , Warm Ischemia/adverse effects
14.
Molecules ; 26(12)2021 Jun 09.
Article in English | MEDLINE | ID: mdl-34207886

ABSTRACT

Interleukin-1ß (IL-1ß), a product of the NLRP3 inflammasome, modulates cardiac contractility and diastolic function. We proposed that OLT1177® (dapansutrile), a novel NLRP3 inhibitor, could preserve contractile reserve and diastolic function after myocardial infarction (MI). We used an experimental murine model of severe ischemic cardiomyopathy through the ligation of the left coronary artery without reperfusion, and after 7 days randomly assigned mice showing large anterior MI (>4 akinetic segments), increased left ventricular (LV) dimensions ([LVEDD] > 4.4 mm), and reduced function (LV ejection fraction < 40%) to a diet that was enriched with OLT1177® admixed with the chow in the diet at 3.75 g/kg (Group 1 [n = 10]) or 7.5 g/kg (Group 2 [n = 9]), or a standard diet as the no-treatment control group (Group 3 [n = 10]) for 9 weeks. We measured the cardiac function and contractile reserve with an isoproterenol challenge, and the diastolic function with cardiac catheterization at 10 weeks following the MI surgery. When compared with the control (Group 3), the mice treated with OLT1177 (Group 1 and 2) showed significantly greater preservation of their contractile reserve (the percent increase in the left ventricular ejection fraction [LVEF] after the isoproterenol challenge was +33 ± 11% and +40 ± 6% vs. +9 ± 7% in the standard diet; p < 0.05 and p < 0.005 for Group 1 and 2, respectively) and of diastolic function measured as the lower left ventricular end-diastolic pressure (3.2 ± 0.5 mmHg or 4.5 ± 0.5 mmHg vs. 10.0 ± 1.6 mmHg; p < 0.005 and p < 0.009 respectively). No differences were noted between the resting LVEF of the MI groups. These effects were independent of the effects on the ventricular remodeling after MI. NLRP3 inflammasome inhibition with OLT1177® can preserve ß-adrenergic responsiveness and prevent left ventricular diastolic dysfunction in a large non-reperfused anterior MI mouse model. OLT1177® could therefore be used to prevent the development of heart failure in patients with ischemic cardiomyopathy.


Subject(s)
Anterior Wall Myocardial Infarction/drug therapy , Cardiomyopathies/drug therapy , Myocardial Ischemia/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Nitriles/pharmacology , Animals , Anterior Wall Myocardial Infarction/metabolism , Anterior Wall Myocardial Infarction/pathology , Anti-Inflammatory Agents/pharmacology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Diastole , Disease Models, Animal , Inflammasomes/antagonists & inhibitors , Male , Mice , Mice, Inbred ICR , Myocardial Contraction , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology
15.
Nanoscale Res Lett ; 16(1): 118, 2021 Jul 22.
Article in English | MEDLINE | ID: mdl-34292415

ABSTRACT

In this work, we used a sequential method of synthesis for gold-silver bimetallic nanoparticles with core@shell structure (Au@AgNPs). Rumex hymenosepalus root extract (Rh), which presents high content in catechins and stilbenes, was used as reductor agent in nanoparticles synthesis. Size distribution obtained by Transmission Electron Microscopy (TEM) gives a mean diameter of 36 ± 11 nm for Au@AgNPs, 24 ± 4 nm for gold nanoparticles (AuNPs), and 13 ± 3 nm for silver nanoparticles (AgNPs). The geometrical shapes of NPs were principally quasi-spherical. The thickness of the silver shell over AuNPs is around 6 nm and covered by active biomolecules onto the surface. Nanoparticles characterization included high angle annular dark field images (HAADF) recorded with a scanning transmission electron microscope (STEM), Energy-Dispersive X-ray Spectroscopy (EDS), X-Ray Diffraction (XRD), UV-Vis Spectroscopy, Zeta Potential, and Dynamic Light Scattering (DLS). Fourier Transform Infrared Spectrometer (FTIR), and X-ray Photoelectron Spectroscopy (XPS) show that nanoparticles are stabilized by extract molecules. A growth kinetics study was performed using the Gompertz model for microorganisms exposed to nanomaterials. The results indicate that AgNPs and Au@AgNPs affect the lag phase and growth rate of Escherichia coli and Candida albicans in a dose-dependent manner, with a better response for Au@AgNPs.

16.
Sci Rep ; 11(1): 11312, 2021 05 28.
Article in English | MEDLINE | ID: mdl-34050228

ABSTRACT

In this work we use Mimosa tenuiflora (MtE) extracts as reducing agents to synthesize silver nanoparticles (AgMt NPs) which were characterized by DPPH and Total Polyphenols Assays, UV-visible, X-ray diffractometer (XRD), high-resolution transmission electron microscopy (HRTEM), X-ray photoelectron spectroscopy (XPS) and Thermogravimetric analysis (TGA). AgMt NPs possess average sizes of 21 nm and fcc crystalline structure, it was also confirmed that the MtE is present in the AgMt NPs even after the cleaning protocol applied. Subsequently, carbopol hydrogels were made and the MtE and the synthesized AgMt NPs were dispersed in different gels (MtE-G and AgMt NPs-G, respectively) at 100 µg/g concentration. The gels were characterized by UV-Vis, IR, and rheology. Antimicrobial tests were performed using Staphylococcus aureus and Escherichia coli. Burn wound healing was evaluated in a second-degree burn injury on a Wistar rats model for 14 days and additional skin biopsies were examined with histopathological analysis. Gel with commercial silver nanoparticles (Ag NPs) was prepared and employed as a control on the biological assays. Hydrogel system containing silver nanoparticles synthesized with Mimosa tenuiflora (AgMt NPs-G) is a promising therapeutic strategy for burn wound healing, this due to bactericidal and anti-inflammatory effects, which promotes a more effective recovery (in percentage terms) by damaged area.


Subject(s)
Burns/drug therapy , Mimosa/chemistry , Plant Extracts/administration & dosage , Silver/administration & dosage , Animals , Drug Evaluation, Preclinical , Hydrogels/chemistry , Hydrogels/therapeutic use , Male , Metal Nanoparticles/chemistry , Metal Nanoparticles/ultrastructure , Phytotherapy , Plant Bark/chemistry , Plant Extracts/chemistry , Rats, Wistar , Silver/chemistry , Wound Healing/drug effects
17.
Radiat Res ; 195(5): 463-473, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33822229

ABSTRACT

After radiation exposure, endothelium-dependent vasorelaxation is impaired due to impaired nitric oxide production. Endothelial dysfunction is characterized by uncoupled endothelial nitric oxide synthase activity, oxidation of the reduced cofactor tetrahydrobiopterin to dihydrobiopterin as one well recognized mechanism. Oral treatment with sepiapterin, a tetrahydrobiopterin precursor, decreased infiltrating inflammatory cells and cytokine levels in mice with colitis. We therefore tested whether a synthetic sepiapterin, PTC923, might mitigate radiation-induced cardiac and pulmonary injuries. C57L/J wild-type 6-8-week-old mice of both sexes received 5 Gy total-body irradiation (TBI), followed by a top-up dose of 6.5 Gy to the thorax (total thoracic dose of 11.5 Gy). Starting from 24 h postirradiation, mice were treated once daily with 1 mg/kg PTC923 for six days by oral gavage. Assessment of lung injury by breathing rate was measured every other week and echocardiography to assess heart function was performed at different time points (8, 30, 60, 90 and 180 days). Plasma proteins (fibrinogen, neutrophil elastase, C-reactive protein, and IL-6) were assessed as well. TBI induced a reduction in cardiac contractile reserve and an impairment in diastolic function restored by daily oral PTC923. Postirradiation lung injury was significantly delayed by PTC923. TBI mice treated with PTC923 experienced a longer survival compared to nonirradiated mice (71% vs. 40% of mice alive after 180 days). PTC923-treated mice showed a reduction in inflammatory mediators, especially IL-6 and IL-1b. In conclusion, these findings support the proposal that PTC923 is a potential mitigator of cardiac and lung injury caused by TBI.


Subject(s)
Heart Injuries/drug therapy , Heart Injuries/etiology , Lung Injury/drug therapy , Lung Injury/etiology , Pterins/administration & dosage , Pterins/pharmacology , Whole-Body Irradiation/adverse effects , Administration, Oral , Animals , Dose-Response Relationship, Radiation , Female , Heart Injuries/metabolism , Inflammation Mediators/metabolism , Lung Injury/metabolism , Male , Mice , Mice, Inbred C57BL , Pterins/therapeutic use , Time Factors
18.
JACC Basic Transl Sci ; 6(2): 137-150, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33665514

ABSTRACT

Human samples of patients with chronic pericarditis and appropriate control subjects were stained for the inflammasome components. A mouse model of pericarditis was developed through the intrapericardial injection of zymosan A. Different inflammasome blockers were tested in the mouse model. Patients with pericarditis presented an intensification of the inflammasome activation compared with control subjects. The experimental model showed the pathological features of pericarditis. Among inflammasome blockers, NLRP3 inflammasome inhibitor, anakinra, and interleukin-1 trap were found to significantly improve pericardial alterations. Colchicine partially improved the pericardial inflammation. An intense activation of the inflammasome in pericarditis was demonstrated both in humans and in mice.

19.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33649199

ABSTRACT

Interleukin-1ß (IL-1ß)-mediated inflammation suppresses antitumor immunity, leading to the generation of a tumor-permissive environment, tumor growth, and progression. Here, we demonstrate that nucleotide-binding domain, leucine-rich containing family, pyrin domain-containing-3 (NLRP3) inflammasome activation in melanoma is linked to IL-1ß production, inflammation, and immunosuppression. Analysis of cancer genome datasets (TCGA and GTEx) revealed greater NLRP3 and IL-1ß expression in cutaneous melanoma samples (n = 469) compared to normal skin (n = 324), with a highly significant correlation between NLRP3 and IL-1ß (P < 0.0001). We show the formation of the NLRP3 inflammasome in biopsies of metastatic melanoma using fluorescent resonance energy transfer analysis for NLRP3 and apoptosis-associated speck-like protein containing a CARD. In vivo, tumor-associated NLRP3/IL-1 signaling induced expansion of myeloid-derived suppressor cells (MDSCs), leading to reduced natural killer and CD8+ T cell activity concomitant with an increased presence of regulatory T (Treg) cells in the primary tumors. Either genetic or pharmacological inhibition of tumor-derived NLRP3 by dapansutrile (OLT1177) was sufficient to reduce MDSCs expansion and to enhance antitumor immunity, resulting in reduced tumor growth. Additionally, we observed that the combination of NLRP3 inhibition and anti-PD-1 treatment significantly increased the antitumor efficacy of the monotherapy by limiting MDSC-mediated T cell suppression and tumor progression. These data show that NLRP3 activation in melanoma cells is a protumor mechanism, which induces MDSCs expansion and immune evasion. We conclude that inhibition of NLRP3 can augment the efficacy of anti-PD-1 therapy.


Subject(s)
Melanoma, Experimental/immunology , Myeloid-Derived Suppressor Cells/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , Neoplasm Proteins/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Neoplasm Proteins/genetics , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology
20.
Inflamm Res ; 70(1): 7-10, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33079210

ABSTRACT

OBJECTIVE: The orf8b protein of the coronavirus SARS-CoV, analogous to SARS-CoV-2, triggers the NLRP3 inflammasome in macrophages in vitro. Deregulated inflammasome-mediated release of interleukin-1 family cytokines is important in hyper-inflammatory syndromes, like happens in SARS-CoV-2-mediated cytokine release syndrome. We propose that an intense inflammasome formation characterizes the lungs of patients with fatal COVID-19 disease due to pneumonia and acute respiratory distress syndrome (ARDS). METHODS: Samples from four patients with confirmed COVID-19 pneumonia who had been hospitalized at the Hospital of the University of Trieste (Italy) and died of ARDS and four lung samples from a historical repository from subjects who had died of cardiopulmonary arrest and had not been placed on mechanical ventilation and without evidence of pulmonary infection at postmortem examination were collected. Pathology samples had been fixed in formalin 10% at time of collection and subsequently embedded in paraffin. We conducted staining for ASC (Apoptosis-associated Speck-like protein containing a Caspase recruitment domain), NLRP3 (NACHT, LRR, and PYD domains-containing protein 3), and cleaved caspase-1. RESULTS: Intense expression of the inflammasome was detected, mainly in leukocytes, within the lungs of all patients with fatal COVID-19 in the areas of lung injury. The number of ASC inflammasome specks per high power fields was significantly higher in the lungs of patients with fatal COVID-19 as compared with the lungs of control subjects (52 ± 22 vs 6 ± 3, P = 0.0064). CONCLUSIONS: These findings identify the presence of NLRP3 inflammasome aggregates in the lungs of fatal COVID-19 pneumonia thus providing the potential molecular link between viral infection and cytokine release syndrome.


Subject(s)
COVID-19/pathology , Inflammasomes , Lung/pathology , Adult , Aged , Autopsy , CARD Signaling Adaptor Proteins/analysis , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/analysis , Caspase 1/metabolism , Cytokine Release Syndrome/metabolism , Cytokine Release Syndrome/pathology , Female , Heart Arrest/etiology , Humans , Leukocytes/pathology , Male , Middle Aged , NLR Family, Pyrin Domain-Containing 3 Protein/analysis , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pneumonia, Viral/etiology , Pneumonia, Viral/pathology , Respiratory Distress Syndrome/etiology , Respiratory Distress Syndrome/pathology
SELECTION OF CITATIONS
SEARCH DETAIL