Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Perfusion ; 38(7): 1418-1427, 2023 10.
Article in English | MEDLINE | ID: mdl-35849687

ABSTRACT

BACKGROUND: We have previously shown that remote ischemic preconditioning (RIP), which utilizes in part the extracellular RNA (eRNA)/RNase1 pathway, can induce ischemic tolerance in humans. Because RIP has thus far been tested only with four cycles of extremity ischemia/reperfusion, we investigated the influence of six cycles of ischemia on the eRNA/RNase1 pathway in cardiac patients. METHODS: Six cycles of RIP were carried out in 14 patients undergoing cardiac surgery. Blood samples were taken at 13 timepoints during surgery and at three timepoints after surgery for determining serum levels of RNase1, eRNA, and TNF-α. Trans-cardiac gradients between the myocardial blood inflow and outflow were calculated. RESULTS: Between the fourth and the sixth RIP cycles, a noticeable increase in the levels of eRNA (fourth: 151.6 (SD: 44.2) ng/ml vs sixth: 181.8 (SD: 87.5) ng/ml, p = .071), and a significant increase in RNase1 (fourth: 151.1 (SD: 42.6) U/ml vs sixth: 175.3 (SD: 41.2) U/ml, p = .001), were noted. The trans-cardiac gradients of RNase1 and eRNA before and after ischemia were not significantly different (p = .158 and p = .221; p = .397 and p = .683, respectively). Likewise, the trans-cardiac gradient of TNF-α was similar before and after ischemia. During the first 48 h after the surgery, RNase1 activity rose significantly and exceeded baseline values (135.7 (SD: 40.6) U/ml before and 279.2 (SD: 85.6) U/ml after surgery, p = .001) as did eRNA levels (148,6 (SD: 35.4) ng/ml before and 396.5 (SD: 154.5) ng/ml after surgery, p = .005), whereas TNF-α levels decreased significantly (91.7 (SD: 47.7) pg/ml before and 35.7 (SD: 36.9) pg/ml after surgery, p = .001). CONCLUSION: Six RIP cycles increased the RNase1 levels significantly above those observed with four cycles. More clinical data are required to show whether this translates into a benefit for patients.


Subject(s)
Cardiac Surgical Procedures , Ischemic Preconditioning , Humans , Tumor Necrosis Factor-alpha/metabolism , Ischemia , Myocardium/metabolism
2.
Discoveries (Craiova) ; 11(3): e174, 2023.
Article in English | MEDLINE | ID: mdl-38405426

ABSTRACT

BACKGROUND: Platelet Inhibition and Clinical Outcomes (PLATO) was a multicenter, randomized double-blind trial assessing efficacy and safety of ticagrelor versus clopidogrel in patients with acute coronary syndrome. The reported mortality benefit of ticagrelor in the PLATO trial has been challenged for over decade, and never confirmed in later trials. OBJECTIVE: To compare if there were any differences when deaths were reported to the FDAby the sponsors or by independent Contract Research Organizations (CRO). METHODS: We obtained the complete PLATO deaths dataset reported to the FDA and revealed that some events were inaccurately reported favoring ticagrelor. The entire FDA list contains precisely detailed 938 PLATO deaths. The CRO reported outcomes from the USA, Russia, Georgia, and most of Ukraine, while sites in 39 other countries were controlled by the trial sponsors. We compared vascular- (code "11"), non-vascular- (code "12"), and unknown (code "97") deaths triaged by the reporting source. RESULTS: Overall, most PLATO deaths were vascular (n=677), less non-vascular (n=159) andunexpectedly many of "other" (n=7) or "unknown" (n=95) origin reported either by sponsors (n=807) or CRO (n=131). The trial sponsors reported more clopidogrel deaths from vascular (313 vs.239), non-vascular (86 vs.58) and unknown (53 vs. 26) causes.In contrast, CRO-monitored sites reported significantly (72 vs. 53; p<0.01) more ticagrelordeaths than after clopidogrel from vascular (51 vs.39), non-vascular (8 vs.7) and unknown (10 vs. 4) causes. CONCLUSION: Deaths were reported differently by sponsors and CRO within the same trial. Since some deaths were misreported by PLATO sponsors, only the CRO data seems mostly reliable. Among all countries, the CRO - reported PLATO-USA outcomes represent the largest and most realistic dataset of realistic evidence suggesting ticagrelor inferiority to clopidogrel for all primary endpoint components including vascular death.

3.
Int J Mol Sci ; 23(16)2022 Aug 12.
Article in English | MEDLINE | ID: mdl-36012303

ABSTRACT

Double-stranded RNA adenosine deaminase 1 (ADAR1) is significantly down-regulated in fibroblasts derived from Idiopathic Pulmonary Fibrosis (IPF) patients, and its overexpression restored levels of miRNA-21, PELI1, and SPRY2. There are two ADAR1 isoforms in humans, ADAR1-p110 and ADAR1-p150, generated by an alternative promoter. Let-7d is considered an essential microRNA in Pulmonary Fibrosis (PF). In silico analysis revealed COL3A1 and SMAD2, proteins involved in the development of IPF, as Let-7d targets. We analyzed the role of ADAR1-p110 and ADAR1-p150 isoforms in the regulation of Let-7d maturation and the effect of this regulation on the expression of COL3A1 and SMAD2 in IPF fibroblast. We demonstrated that differential expression and subcellular distribution of ADAR1 isoforms in fibroblasts contribute to the up-regulation of pri-miR-Let-7d and down-regulation of mature Let-7d. Induction of overexpression of ADAR1 reestablishes the expression of pri-miR-Let-7d and Let-7d in lung fibroblasts. The reduction of mature Let-7d upregulates the expression of COL3A1 and SMAD2. Thus, ADAR1 isoforms and Let-7d could have a synergistic role in IPF, which is a promising explanation in the mechanisms of fibrosis development, and the regulation of both molecules could be used as a therapeutic approach in IPF.


Subject(s)
Adenosine Deaminase , Idiopathic Pulmonary Fibrosis , MicroRNAs , Adenosine Deaminase/genetics , Adenosine Deaminase/metabolism , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Lung/metabolism , Membrane Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA-Binding Proteins
4.
Nat Commun ; 12(1): 1072, 2021 02 16.
Article in English | MEDLINE | ID: mdl-33594057

ABSTRACT

In addition to nucleosomes, chromatin contains non-histone chromatin-associated proteins, of which the high-mobility group proteins are the most abundant. Chromatin-mediated regulation of transcription involves DNA methylation and histone modifications. However, the order of events and the precise function of high-mobility group proteins during transcription initiation remain unclear. Here we show that high-mobility group AT-hook 2 protein (HMGA2) induces DNA nicks at the transcription start site, which are required by the histone chaperone FACT complex to incorporate nucleosomes containing the histone variant H2A.X. Further, phosphorylation of H2A.X at S139 (γ-H2AX) is required for repair-mediated DNA demethylation and transcription activation. The relevance of these findings is demonstrated within the context of TGFB1 signaling and idiopathic pulmonary fibrosis, suggesting therapies against this lethal disease. Our data support the concept that chromatin opening during transcriptional initiation involves intermediates with DNA breaks that subsequently require DNA repair mechanisms to ensure genome integrity.


Subject(s)
DNA Demethylation , Nucleosomes/metabolism , Transcription Initiation, Genetic , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Chromatin/chemistry , Chromatin/metabolism , HEK293 Cells , HMGA2 Protein/metabolism , Histones/metabolism , Humans , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Mice , Phosphorylation , Phosphoserine/metabolism , RNA Polymerase II/metabolism , Transcription Initiation Site , Transcriptional Activation/genetics , Transforming Growth Factor beta1/metabolism
5.
Am J Ther ; 27(6): e563-e572, 2020.
Article in English | MEDLINE | ID: mdl-33109913

ABSTRACT

BACKGROUND: Excess vascular deaths in the PLATO trial comparing ticagrelor to clopidogrel have been repeatedly challenged by the Food and Drug Administration (FDA) reviewers and academia. Based on the Freedom of Information Act, BuzzFeed won a court order and shared with us the complete list of reported deaths for the ticagrelor FDA New Drug Application (NDA) 22-433. This dataset was matched against local patient-level records from PLATO sites monitored by the sponsor. STUDY QUESTION: Whether FDA death data in the PLATO trial matched the local site records. STUDY DESIGN: The NDA spreadsheet contains 938 precisely detailed PLATO deaths. We obtained and validated local evidence for 52 deaths among 861 PLATO patients from 14 enrolling sites in 8 countries and matched those with the official NDA dataset submitted to the FDA. MEASURES AND OUTCOMES: Existence, precise time, and primary cause of deaths in PLATO. RESULTS: Discrepant to the NDA document, sites confirmed 2 extra unreported deaths (Poland and Korea) and failed to confirm 4 deaths (Malaysia). Of the remaining 46 deaths, dates were reported correctly for 42 patients, earlier (2 clopidogrel), or later (2 ticagrelor) than the actual occurrence of death. In 12 clopidogrel patients, cause of death was changed to "vascular," whereas 6 NDA ticagrelor "nonvascular" or "unknown" deaths were site-reported as of "vascular" origin. Sudden death was incorrectly reported in 4 clopidogrel patients, but omitted in 4 ticagrelor patients directly affecting the primary efficacy PLATO endpoint. CONCLUSIONS: Many deaths were inaccurately reported in PLATO favoring ticagrelor. The full extent of mortality misreporting is currently unclear, while especially worrisome is a mismatch in identifying primary death cause. Because all PLATO events are kept in the cloud electronic Medidata Rave capture system, securing the database content, examining the dataset changes or/and repeated entries, identifying potential interference origin, and assessing full magnitude of the problem are warranted.


Subject(s)
Acute Coronary Syndrome/mortality , Cause of Death , Data Accuracy , Platelet Aggregation Inhibitors/administration & dosage , Acute Coronary Syndrome/complications , Acute Coronary Syndrome/drug therapy , Clinical Trials, Phase III as Topic , Clopidogrel/administration & dosage , Clopidogrel/adverse effects , Datasets as Topic , Drug Approval , Humans , Platelet Aggregation Inhibitors/adverse effects , Randomized Controlled Trials as Topic , Ticagrelor/administration & dosage , Ticagrelor/adverse effects , Treatment Outcome , United States , United States Food and Drug Administration/legislation & jurisprudence , United States Food and Drug Administration/standards
7.
Arch Med Res ; 51(3): 268-277, 2020 04.
Article in English | MEDLINE | ID: mdl-32143939

ABSTRACT

BACKGROUND: There is evidence that the pharmacokinetics of certain drugs in Mexicans may differ with respect to other ethnic groups. On the other hand, there is controversy about the existence of interethnic variability in the pharmacokinetics of ciprofloxacin. AIM OF THE STUDY: To study oral ciprofloxacin pharmacokinetics in Mexicans at various dose levels and make comparisons with other populations in order to gain insight on interethnic variability. METHODS: Healthy Mexican volunteers received oral ciprofloxacin as 250 mg and 500 mg immediate-release tablets or a 1,000 mg extended-release formulation. Plasma concentration against time curves were constructed, and pharmacokinetic parameters were compared with those reported for other populations. RESULTS: Ciprofloxacin pharmacokinetics in Mexicans was linear and no significant differences between males and females were detected. When several populations were compared, it appeared that bioavailability in Mexicans was similar to that of Caucasians, being lower than that of Asians. These variations were attenuated when data were normalized by body weight. CONCLUSIONS: Ciprofloxacin pharmacokinetics exhibit interethnic variability, Asians exhibiting an increased bioavailability with regard to Mexicans and Caucasians. Data suggest that these differences are due to body weight.


Subject(s)
Ciprofloxacin/blood , Ciprofloxacin/pharmacokinetics , Healthy Volunteers/statistics & numerical data , Administration, Oral , Adult , Asian People , Biological Availability , Body Weight/physiology , Ethnicity , Female , Humans , Male , Mexico , White People , Young Adult
8.
J Cell Mol Med ; 24(7): 3795-3806, 2020 04.
Article in English | MEDLINE | ID: mdl-32155321

ABSTRACT

Acute myocardial infarction causes lethal injury to cardiomyocytes during both ischaemia and reperfusion (IR). It is important to define the precise mechanisms by which they die in order to develop strategies to protect the heart from IR injury. Necrosis is known to play a major role in myocardial IR injury. There is also evidence for significant myocardial death by other pathways such as apoptosis, although this has been challenged. Mitochondria play a central role in both of these pathways of cell death, as either a causal mechanism is the case of mitochondrial permeability transition leading to necrosis, or as part of the signalling pathway in mitochondrial cytochrome c release and apoptosis. Autophagy may impact this process by removing dysfunctional proteins or even entire mitochondria through a process called mitophagy. More recently, roles for other programmed mechanisms of cell death such as necroptosis and pyroptosis have been described, and inhibitors of these pathways have been shown to be cardioprotective. In this review, we discuss both mitochondrial and mitochondrial-independent pathways of the major modes of cell death, their role in IR injury and their potential to be targeted as part of a cardioprotective strategy. This article is part of a special Issue entitled 'Mitochondria as targets of acute cardioprotection' and emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.


Subject(s)
Mitochondria/genetics , Myocardial Infarction/genetics , Myocardial Reperfusion Injury/genetics , Myocardium/metabolism , Apoptosis/genetics , Autophagy/genetics , Cell Death/genetics , Humans , Mitochondria/pathology , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/pathology , Necrosis/genetics , Necrosis/pathology , Signal Transduction/genetics
9.
Indian J Med Res ; 151(1): 11-21, 2020 01.
Article in English | MEDLINE | ID: mdl-32134010

ABSTRACT

Obesity is a serious medical condition worldwide, which needs new approaches and recognized international consensus in treating diseases leading to morbidity. The aim of this review was to examine heterogeneous links among the various phenotypes of obesity in adults. Proteins and associated genes in each group were analysed to differentiate between biomarkers. A variety of terms for classification and characterization within this pathology are currently in use; however, there is no clear consensus in terminology. The most significant groups reviewed include metabolically healthy obese, metabolically abnormal obese, metabolically abnormal, normal weight and sarcopenic obese. These phenotypes do not define particular genotypes or epigenetic gene regulation, or proteins related to inflammation. There are many other genes linked to obesity, though the value of screening all of those for diagnosis has low predictive results, as there are no significant biomarkers. It is important to establish a consensus in the terminology used and the characteristics attributed to obesity subtypes. The identification of specific molecular biomarkers is also required for better diagnosis in subtypes of obesity.


Subject(s)
Biomarkers , Obesity/diagnosis , Obesity/genetics , Proteins/genetics , Adult , Genotype , Humans , Obesity/classification , Obesity/epidemiology , Phenotype
10.
Br J Pharmacol ; 177(23): 5357-5374, 2020 12.
Article in English | MEDLINE | ID: mdl-31943142

ABSTRACT

The association between chronic inflammatory diseases (CIDs) and increased cardiovascular (CV) risk is well documented and can be a most threatening complication in these patients. However, the pathogenetic mechanisms underlying increased CV risk remain elusive, especially in their cellular and biochemical pathways. Using animal models to understand mechanisms underlying cardiac involvement are limited. Additionally, treatments may influence cardiovascular events through different outcomes. Some drugs used to treat CIDs can negatively affect cardiac function by a direct toxicity, whereas others may protect the myocardium. In the present article, we focus on the cardiac manifestations and risk factors, the pathogenetic mechanisms, and the effect of treatments on myocardial function and cardioprotection for five common worldwide CIDs (rheumatoid arthritis, systemic lupus erythematosus, systemic sclerosis, psoriasis and inflammatory bowel disease). We also give recommendations in order to evaluate common targets between CID and CV disease (CVD) and to design therapies to alleviate CID-related CVD. LINKED ARTICLES: This article is part of a themed issue on Risk factors, comorbidities, and comedications in cardioprotection. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.23/issuetoc.


Subject(s)
Cardiomyopathies , Cardiovascular Diseases , Animals , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/prevention & control , Chronic Disease , Humans , Myocardium
11.
J Cell Mol Med ; 24(5): 2717-2729, 2020 03.
Article in English | MEDLINE | ID: mdl-31967733

ABSTRACT

Pre-clinical studies have indicated that mitoprotective drugs may add cardioprotection beyond rapid revascularization, antiplatelet therapy and risk modification. We review the clinical efficacy of mitoprotective drugs that have progressed to clinical testing comprising cyclosporine A, KAI-9803, MTP131 and TRO 40303. Whereas cyclosporine may reduce infarct size in patients undergoing primary angioplasty as evaluated by release of myocardial ischaemic biomarkers and infarct size imaging, the other drugs were not capable of demonstrating this effect in the clinical setting. The absent effect leaves the role of the mitochondrial permeability transition pore for reperfusion injury in humans unanswered and indicates that targeting one single mechanism to provide mitoprotection may not be efficient. Moreover, the lack of effect may relate to favourable outcome with current optimal therapy, but conditions such as age, sex, diabetes, dyslipidaemia and concurrent medications may also alter mitochondrial function. However, as long as the molecular structure of the pore remains unknown and specific inhibitors of its opening are lacking, the mitochondrial permeability transition pore remains a target for alleviation of reperfusion injury. Nevertheless, taking conditions such as ageing, sex, comorbidities and co-medication into account may be of paramount importance during the design of pre-clinical and clinical studies testing mitoprotective drugs.


Subject(s)
Mitochondria, Heart/pathology , Myocardial Reperfusion , Protective Agents/therapeutic use , ST Elevation Myocardial Infarction/drug therapy , Translational Research, Biomedical , Animals , Clinical Trials as Topic , Humans , Protective Agents/pharmacology , ST Elevation Myocardial Infarction/physiopathology
12.
Cond Med ; 3(1): 18-30, 2020 Feb.
Article in English | MEDLINE | ID: mdl-34268485

ABSTRACT

Acute myocardial infarction (AMI), and the heart failure (HF) that often follows, are leading causes of death and disability worldwide. Crucially, there are currently no effective treatments, other than myocardial reperfusion, for reducing myocardial infarct (MI) size and preventing HF following AMI. Thus, there is an unmet need to discover novel cardioprotective therapies to reduce MI size, and prevent HF in AMI patients. Although a large number of therapies have been shown to reduce MI size in experimental studies, the majority have failed to benefit AMI patients. Failure to deliver cardioprotective therapy to the ischemic heart in sufficient concentrations following AMI is a major factor for the lack of success observed in previous clinical cardioprotection studies. Therefore, new strategies are needed to improve the delivery of cardioprotective therapies to the ischemic heart following AMI. In this regard, nanoparticles have emerged as drug delivery systems for improving the bioavailability, delivery, and release of cardioprotective therapies, and should result in improved efficacy in terms of reducing MI size and preventing HF. In this article, we provide a review of currently available nanoparticles, some of which have been FDA-approved, in terms of their use as drug delivery systems in cardiovascular disease and cardioprotection.

13.
Int J Mol Sci ; 20(16)2019 Aug 15.
Article in English | MEDLINE | ID: mdl-31443187

ABSTRACT

Background: New treatments are needed to reduce myocardial infarct size (MI) and prevent heart failure (HF) following acute myocardial infarction (AMI), which are the leading causes of death and disability worldwide. Studies in rodent AMI models showed that genetic and pharmacological inhibition of mitochondrial fission, induced by acute ischemia and reperfusion, reduced MI size. Whether targeting mitochondrial fission at the onset of reperfusion is also cardioprotective in a clinically-relevant large animal AMI model remains to be determined. Methods: Adult pigs (30-40 kg) were subjected to closed-chest 90-min left anterior descending artery ischemia followed by 72 h of reperfusion and were randomized to receive an intracoronary bolus of either mdivi-1 (1.2 mg/kg, a small molecule inhibitor of the mitochondrial fission protein, Drp1) or vehicle control, 10-min prior to reperfusion. The left ventricular (LV) size and function were both assessed by transthoracic echocardiography prior to AMI and after 72 h of reperfusion. MI size and the area-at-risk (AAR) were determined using dual staining with Tetrazolium and Evans blue. Heart samples were collected for histological determination of fibrosis and for electron microscopic analysis of mitochondrial morphology. Results: A total of 14 pigs underwent the treatment protocols (eight control and six mdivi-1). Administration of mdivi-1 immediately prior to the onset of reperfusion did not reduce MI size (MI size as % of AAR: Control 49.2 ± 8.6 vs. mdivi-1 50.5 ± 11.4; p = 0.815) or preserve LV systolic function (LV ejection fraction %: Control 67.5 ± 0.4 vs. mdivi-1 59.6 ± 0.6; p = 0.420), when compared to vehicle control. Similarly, there were no differences in mitochondrial morphology or myocardial fibrosis between mdivi-1 and vehicle control groups. Conclusion: Our pilot study has shown that treatment with mdivi-1 (1.2 mg/kg) at the onset of reperfusion did not reduce MI size or preserve LV function in the clinically-relevant closed-chest pig AMI model. A larger study, testing different doses of mdivi-1 or using a more specific Drp1 inhibitor are required to confirm these findings.


Subject(s)
Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Quinazolinones/therapeutic use , Animals , Disease Models, Animal , Echocardiography , Female , Mitochondrial Dynamics/drug effects , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Pilot Projects , Swine , Ventricular Function, Left/drug effects
14.
Nat Commun ; 10(1): 2229, 2019 05 20.
Article in English | MEDLINE | ID: mdl-31110176

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and highly lethal lung disease with unknown etiology and poor prognosis. IPF patients die within 2 years after diagnosis mostly due to respiratory failure. Current treatments against IPF aim to ameliorate patient symptoms and to delay disease progression. Unfortunately, therapies targeting the causes of or reverting IPF have not yet been developed. Here we show that reduced levels of miRNA lethal 7d (MIRLET7D) in IPF compromise epigenetic gene silencing mediated by the ribonucleoprotein complex MiCEE. In addition, we find that hyperactive EP300 reduces nuclear HDAC activity and interferes with MiCEE function in IPF. Remarkably, EP300 inhibition reduces fibrotic hallmarks of in vitro (patient-derived primary fibroblast), in vivo (bleomycin mouse model), and ex vivo (precision-cut lung slices, PCLS) IPF models. Our work provides the molecular basis for therapies against IPF using EP300 inhibition.


Subject(s)
E1A-Associated p300 Protein/metabolism , Histone Deacetylase 1/metabolism , Idiopathic Pulmonary Fibrosis/pathology , MicroRNAs/metabolism , Ribonucleoproteins/metabolism , Animals , Bleomycin/toxicity , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cells, Cultured , Disease Models, Animal , E1A-Associated p300 Protein/antagonists & inhibitors , Fibroblasts , Gene Silencing , Histone Deacetylase 2/metabolism , Humans , Idiopathic Pulmonary Fibrosis/chemically induced , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/genetics , Lung/drug effects , Lung/pathology , Male , Mice , Primary Cell Culture , Ribonucleoproteins/genetics
15.
Int J Mol Sci ; 20(10)2019 May 19.
Article in English | MEDLINE | ID: mdl-31109146

ABSTRACT

Cardiovascular diseases are the leading cause of mortality worldwide. It is widely known that non-resolving inflammation results in atherosclerotic conditions, which are responsible for a host of downstream pathologies including thrombosis, myocardial infarction (MI), and neurovascular events. Macrophages, as part of the innate immune response, are among the most important cell types in every stage of atherosclerosis. In this review we discuss the principles governing macrophage function in the healthy and infarcted heart. More specifically, how cardiac macrophages participate in myocardial infarction as well as cardiac repair and remodeling. The intricate balance between phenotypically heterogeneous populations of macrophages in the heart have profound and highly orchestrated effects during different phases of myocardial infarction. In the early "inflammatory" stage of MI, resident cardiac macrophages are replaced by classically activated macrophages derived from the bone marrow and spleen. And while the macrophage population shifts towards an alternatively activated phenotype, the inflammatory response subsides giving way to the "reparative/proliferative" phase. Lastly, we describe the therapeutic potential of cardiac macrophages in the context of cell-mediated cardio-protection. Promising results demonstrate innovative concepts; one employing a subset of yolk sac-derived, cardiac macrophages that have complete restorative capacity in the injured myocardium of neonatal mice, and in another example, post-conditioning of cardiac macrophages with cardiosphere-derived cells significantly improved patient's post-MI diagnoses.


Subject(s)
Cardiovascular Diseases/immunology , Immunity, Innate , Macrophages/immunology , Myocardium/immunology , Animals , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/therapy , Heart/physiopathology , Humans , Macrophages/pathology , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Myocardium/cytology , Myocardium/pathology , Protective Factors , Ventricular Remodeling
16.
Cardiovasc Res ; 115(7): 1117-1130, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30825305

ABSTRACT

New therapies are required to reduce myocardial infarct (MI) size and prevent the onset of heart failure in patients presenting with acute myocardial infarction (AMI), one of the leading causes of death and disability globally. In this regard, the immune cell response to AMI, which comprises an initial pro-inflammatory reaction followed by an anti-inflammatory phase, contributes to final MI size and post-AMI remodelling [changes in left ventricular (LV) size and function]. The transition between these two phases is critical in this regard, with a persistent and severe pro-inflammatory reaction leading to adverse LV remodelling and increased propensity for developing heart failure. In this review article, we provide an overview of the immune cells involved in orchestrating the complex and dynamic inflammatory response to AMI-these include neutrophils, monocytes/macrophages, and emerging players such as dendritic cells, lymphocytes, pericardial lymphoid cells, endothelial cells, and cardiac fibroblasts. We discuss potential reasons for past failures of anti-inflammatory cardioprotective therapies, and highlight new treatment targets for modulating the immune cell response to AMI, as a potential therapeutic strategy to improve clinical outcomes in AMI patients. This article is part of a Cardiovascular Research Spotlight Issue entitled 'Cardioprotection Beyond the Cardiomyocyte', and emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cardiovascular Agents/therapeutic use , Heart Failure/prevention & control , Myocardial Infarction/therapy , Myocardial Reperfusion Injury/prevention & control , Myocardium/immunology , Animals , Anti-Inflammatory Agents/adverse effects , Cardiovascular Agents/adverse effects , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dendritic Cells/metabolism , Fibroblasts/drug effects , Fibroblasts/immunology , Fibroblasts/metabolism , Heart Failure/immunology , Heart Failure/metabolism , Heart Failure/pathology , Humans , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Lymphocytes/drug effects , Lymphocytes/immunology , Lymphocytes/metabolism , Mast Cells/drug effects , Mast Cells/immunology , Mast Cells/metabolism , Monocytes/drug effects , Monocytes/immunology , Monocytes/metabolism , Myocardial Infarction/immunology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/metabolism , Signal Transduction , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
17.
Arterioscler Thromb Vasc Biol ; 39(3): 387-401, 2019 03.
Article in English | MEDLINE | ID: mdl-30651003

ABSTRACT

Objective- Atherosclerotic coronary artery disease is the leading cause of death worldwide, and current treatment options are insufficient. Using systems-level network cluster analyses on a large coronary artery disease case-control cohort, we previously identified PCSK3 (proprotein convertase subtilisin/kexin family member 3; FURIN) as a member of several coronary artery disease-associated pathways. Thus, our objective is to determine the role of FURIN in atherosclerosis. Approach and Results- In vitro, FURIN inhibitor treatment resulted in reduced monocyte migration and reduced macrophage and vascular endothelial cell inflammatory and cytokine gene expression. In vivo, administration of an irreversible inhibitor of FURIN, α-1-PDX (α1-antitrypsin Portland), to hyperlipidemic Ldlr-/- mice resulted in lower atherosclerotic lesion area and a specific reduction in severe lesions. Significantly lower lesional macrophage and collagen area, as well as systemic inflammatory markers, were observed. MMP2 (matrix metallopeptidase 2), an effector of endothelial function and atherosclerotic lesion progression, and a FURIN substrate was significantly reduced in the aorta of inhibitor-treated mice. To determine FURIN's role in vascular endothelial function, we administered α-1-PDX to Apoe-/- mice harboring a wire injury in the common carotid artery. We observed significantly decreased carotid intimal thickness and lower plaque cellularity, smooth muscle cell, macrophage, and inflammatory marker content, suggesting protection against vascular remodeling. Overexpression of FURIN in this model resulted in a significant 67% increase in intimal plaque thickness, confirming that FURIN levels directly correlate with atherosclerosis. Conclusions- We show that systemic inhibition of FURIN in mice decreases vascular remodeling and atherosclerosis. FURIN-mediated modulation of MMP2 activity may contribute to the atheroprotection observed in these mice.


Subject(s)
Atherosclerosis/prevention & control , Furin/antagonists & inhibitors , Plaque, Atherosclerotic/drug therapy , alpha 1-Antitrypsin/therapeutic use , Animals , Aorta/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Carotid Artery, Common , Disease Progression , Drug Evaluation, Preclinical , Enzyme Induction/drug effects , Furin/genetics , Furin/physiology , Gene Expression Regulation/drug effects , Macrophages/physiology , Male , Matrix Metalloproteinase 2/analysis , Mice , Mice, Inbred C57BL , Monocytes/physiology , Plaque, Atherosclerotic/pathology , Receptors, LDL/deficiency , Tunica Intima/drug effects , Tunica Intima/pathology , Vascular Remodeling , alpha 1-Antitrypsin/pharmacology
18.
Cardiovasc Res ; 115(7): 1131-1142, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30576455

ABSTRACT

Acute obstruction of a coronary artery causes myocardial ischaemia and if prolonged, may result in an ST-segment elevation myocardial infarction (STEMI). First-line treatment involves rapid reperfusion. However, a highly dynamic and co-ordinated inflammatory response is rapidly mounted to repair and remove the injured cells which, paradoxically, can further exacerbate myocardial injury. Furthermore, although cardiac remodelling may initially preserve some function to the heart, it can lead over time to adverse remodelling and eventually heart failure. Since the size of the infarct corresponds to the subsequent risk of developing heart failure, it is important to find ways to limit initial infarct development. In this review, we focus on the role of the innate immune system in the acute response to ischaemia-reperfusion (IR) and specifically its contribution to cell death and myocardial infarction. Numerous danger-associated molecular patterns are released from dying cells in the myocardium, which can stimulate pattern recognition receptors including toll like receptors and NOD-like receptors (NLRs) in resident cardiac and immune cells. Activation of the NLRP3 inflammasome, caspase 1, and pyroptosis may ensue, particularly when the myocardium has been previously aggravated by the presence of comorbidities. Evidence will be discussed that suggests agents targeting innate immunity may be a promising means of protecting the hearts of STEMI patients against acute IR injury. However, the dosing and timing of such agents should be carefully determined because innate immunity pathways may also be involved in cardioprotection. This article is part of a Cardiovascular Research Spotlight Issue entitled 'Cardioprotection Beyond the Cardiomyocyte', and emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cardiovascular Agents/therapeutic use , Heart Failure/prevention & control , Immunity, Innate , Inflammasomes/drug effects , Myocardial Reperfusion Injury/prevention & control , Myocardium/immunology , ST Elevation Myocardial Infarction/therapy , Animals , Anti-Inflammatory Agents/adverse effects , Cardiovascular Agents/adverse effects , Caspase 1/immunology , Caspase 1/metabolism , Caspase Inhibitors/therapeutic use , Heart Failure/immunology , Heart Failure/metabolism , Heart Failure/pathology , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Molecular Targeted Therapy , Myocardial Reperfusion Injury/immunology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardium/metabolism , Myocardium/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , ST Elevation Myocardial Infarction/immunology , ST Elevation Myocardial Infarction/metabolism , ST Elevation Myocardial Infarction/pathology , Signal Transduction
19.
Cardiovasc Res ; 115(7): 1156-1166, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30590395

ABSTRACT

During an ST-elevation myocardial infarction (STEMI), the myocardium undergoes a prolonged period of ischaemia. Reperfusion therapy is essential to minimize cardiac injury but can paradoxically cause further damage. Experimental procedures to limit ischaemia and reperfusion (IR) injury have tended to focus on the cardiomyocytes since they are crucial for cardiac function. However, there is increasing evidence that non-cardiomyocyte resident cells in the heart (as discussed in a separate review in this Spotlight series) as well as circulating cells and factors play important roles in this pathology. For example, erythrocytes, in addition to their main oxygen-ferrying role, can protect the heart from IR injury via the export of nitric oxide bioactivity. Platelets are well-known to be involved in haemostasis and thrombosis, but beyond these roles, they secrete numerous factors including sphingosine-1 phosphate (S1P), platelet activating factor, and cytokines that can all strongly influence the development of IR injury. This is particularly relevant given that most STEMI patients receive at least one type of platelet inhibitor. Moreover, there are large numbers of circulating vesicles in the blood, including microvesicles and exosomes, which can exert both beneficial and detrimental effects on IR injury. Some of these effects are mediated by the transfer of microRNA (miRNA) to the heart. Synthetic miRNA molecules may offer an alternative approach to limiting the response to IR injury. We discuss these and other circulating factors, focussing on potential therapeutic targets relevant to IR injury. Given the prevalence of comorbidities such as diabetes in the target patient population, their influence will also be discussed. This article is part of a Cardiovascular Research Spotlight Issue entitled 'Cardioprotection Beyond the Cardiomyocyte', and emerged as part of the discussions of the European Union (EU)-CARDIOPROTECTION Cooperation in Science and Technology (COST) Action, CA16225.


Subject(s)
Blood Platelets/metabolism , Erythrocytes/metabolism , Extracellular Vesicles/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , ST Elevation Myocardial Infarction/therapy , Animals , Blood Platelets/drug effects , Circulating MicroRNA/blood , Circulating MicroRNA/therapeutic use , Extracellular Vesicles/transplantation , Hemostasis , Humans , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Platelet Aggregation Inhibitors/therapeutic use , ST Elevation Myocardial Infarction/blood , ST Elevation Myocardial Infarction/pathology , ST Elevation Myocardial Infarction/physiopathology , Signal Transduction
20.
Cond Med ; 1(6): 313-318, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30556056

ABSTRACT

Ischemic heart disease (IHD) is one of the leading causes of death and disability worldwide. Platelets, as the main regulators of hemostasis, are major players in acute myocardial ischemia/reperfusion injury (IRI). Additionally, platelets are modified by endogenous cardioprotective strategies such as ischemic preconditioning, postconditioning, and remote ischemic conditioning. In this article, we provide an overview of the functionional role of platelets in acute myocardial IRI, and highlight their potential as targets for cardioprotection to improve health outcomes in patients with IHD.

SELECTION OF CITATIONS
SEARCH DETAIL
...