Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 356
Filtrar
1.
Exp Biol Med (Maywood) ; 249: 10121, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39104790

RESUMEN

Cardiovascular diseases (CVDs) are the leading cause of death worldwide, having become a global public health problem, so the pathophysiological mechanisms and therapeutic strategies of CVDs need further study. Legumain is a powerful enzyme that is widely distributed in mammals and plays an important role in a variety of biological processes. Recent research suggests that legumain is associated with the occurrence and progression of CVDs. In this review, we provide a comprehensive overview of legumain in the pathogenesis of CVDs. The role of legumain in CVDs, such as carotid atherosclerosis, pulmonary hypertension, coronary artery disease, peripheral arterial disease, aortic aneurysms and dissection, is discussed. The potential applications of legumain as a biomarker of these diseases are also explored. By understanding the role of legumain in the pathogenesis of CVDs, we aim to support new therapeutic strategies to prevent or treat these diseases.


Asunto(s)
Enfermedades Cardiovasculares , Cisteína Endopeptidasas , Humanos , Cisteína Endopeptidasas/metabolismo , Enfermedades Cardiovasculares/enzimología , Animales , Biomarcadores/metabolismo
2.
Arterioscler Thromb Vasc Biol ; 44(9): 1960-1974, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38989576

RESUMEN

BACKGROUND: Patients with JAK2V617F-positive myeloproliferative neoplasms (MPNs) and clonal hematopoiesis of indeterminate potential face a significantly elevated risk of cardiovascular diseases. Endothelial cells carrying the JAK2V617F mutation have been detected in many patients with MPN. In this study, we investigated the molecular basis for the high incidence of cardiovascular complications in patients with MPN. METHODS: We investigated the impact of endothelial JAK2V617F mutation on cardiovascular disease development using both transgenic murine models and MPN patient-derived induced pluripotent stem cell lines. RESULTS: Our investigations revealed that JAK2V617F mutant endothelial cells promote cardiovascular diseases under stress, which is associated with endothelial-to-mesenchymal transition and endothelial dysfunction. Importantly, we discovered that inhibiting the endothelial TPO (thrombopoietin) receptor MPL (myeloproliferative leukemia virus oncogene) suppressed JAK2V617F-induced endothelial-to-mesenchymal transition and prevented cardiovascular dysfunction caused by mutant endothelial cells. Notably, the endothelial MPL receptor is not essential for the normal physiological regulation of blood cell counts and cardiac function. CONCLUSIONS: JAK2V617F mutant endothelial cells play a critical role in the development of cardiovascular diseases in JAK2V617F-positive MPNs, and endothelial MPL could be a promising therapeutic target for preventing or ameliorating cardiovascular complications in these patients.


Asunto(s)
Enfermedades Cardiovasculares , Células Endoteliales , Células Madre Pluripotentes Inducidas , Janus Quinasa 2 , Mutación , Trastornos Mieloproliferativos , Receptores de Trombopoyetina , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Receptores de Trombopoyetina/genética , Animales , Humanos , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/enzimología , Trastornos Mieloproliferativos/complicaciones , Trastornos Mieloproliferativos/metabolismo , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Células Endoteliales/patología , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/etiología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/enzimología , Ratones Transgénicos , Transducción de Señal , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Ratones
3.
J Biol Chem ; 298(9): 102284, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35868561

RESUMEN

cGMP-dependent protein kinase (PKG) represents a compelling drug target for treatment of cardiovascular diseases. PKG1 is the major effector of beneficial cGMP signaling which is involved in smooth muscle relaxation and vascular tone, inhibition of platelet aggregation and signaling that leads to cardioprotection. In this study, a novel piperidine series of activators previously identified from an ultrahigh-throughput screen were validated to directly bind partially activated PKG1α and subsequently enhance its kinase activity in a concentration-dependent manner. Compounds from initial optimization efforts showed an ability to activate PKG1α independent of the endogenous activator, cGMP. We demonstrate these small molecule activators mimic the effect of cGMP on the kinetic parameters of PKG1α by positively modulating the KM of the peptide substrate and negatively modulating the apparent KM for ATP with increase in catalytic efficiency, kcat. In addition, these compounds also allosterically modulate the binding affinity of cGMP for PKG1α by increasing the affinity of cGMP for the high-affinity binding site (CNB-A) and decreasing the affinity of cGMP for the low-affinity binding site (CNB-B). We show the mode of action of these activators involves binding to an allosteric site within the regulatory domain, near the CNB-B binding site. To the best of our knowledge, these are the first reported non-cGMP mimetic small molecules shown to directly activate PKG1α. Insights into the mechanism of action of these compounds will enable future development of cardioprotective compounds that function through novel modes of action for the treatment of cardiovascular diseases.


Asunto(s)
Enfermedades Cardiovasculares , Proteína Quinasa Dependiente de GMP Cíclico Tipo I , GMP Cíclico , Piperidinas , Adenosina Trifosfato/metabolismo , Regulación Alostérica/efectos de los fármacos , Sitio Alostérico/efectos de los fármacos , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/enzimología , GMP Cíclico/metabolismo , Proteína Quinasa Dependiente de GMP Cíclico Tipo I/metabolismo , Humanos , Piperidinas/farmacología , Piperidinas/uso terapéutico , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/farmacología
4.
Oxid Med Cell Longev ; 2022: 6194532, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35126817

RESUMEN

Background and Objective. 5-Fluorouracil is one of the most common chemotherapeutic agents used in the treatment of solid tumors. 5-Fluorouracil-associated cardiotoxicity is the second cause of cardiotoxicity induced by chemotherapeutic drugs after anthracyclines. Colchicine is a strong anti-inflammatory drug used to prevent and treat acute gout and treat familial Mediterranean fever. And also, its protective effects on cardiovascular disease have been reported in various studies. The current study is aimed at appraising the effect of colchicine on 5-fluorouracil-induced cardiotoxicity in rats. Methods. Twenty male Wistar rats were divided into four groups as follows: control, 5-fluorouracil, colchicine (5 mg/kg), and 5-fluorouracil+5 mg/kg colchicine. Cardiotoxicity was induced with an intraperitoneal injection of a single dose of 5-fluorouracil (100 mg/kg). The control group received normal saline, and the treatment groups received colchicine with an intraperitoneal injection for 14 days. Findings. 5-Fluorouracil resulted in significant cardiotoxicity represented by an increase in cardiac enzymes, malondialdehyde levels, cyclooxygenase-2 and tumor necrosis factor-alpha expression, cardiac enzymes, and histopathological degenerations. 5-Fluorouracil treatment also decreased body weight, total antioxidant capacity and catalase values, blood cells, and hemoglobin levels. In addition, 5-fluorouracil disrupted electrocardiographic parameters, including increased elevation in the ST segment and increased QRS duration. Treatment with colchicine reduced oxidative stress, cardiac enzymes, histopathological degenerations, and cyclooxygenase-2 expression in cardiac tissue, improved electrocardiographic disorders, and enhanced the number of blood cells and total antioxidant capacity levels. Moreover, body weight loss was hampered after treatment with colchicine. Our results demonstrated that treatment with colchicine significantly improved cardiotoxicity induced by 5-fluorouracil in rats.


Asunto(s)
Antimetabolitos Antineoplásicos/efectos adversos , Antioxidantes/administración & dosificación , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Cardiovasculares/tratamiento farmacológico , Colchicina/administración & dosificación , Colchicum/química , Fluorouracilo/efectos adversos , Fitoquímicos/administración & dosificación , Fitoterapia/métodos , Extractos Vegetales/administración & dosificación , Animales , Antimetabolitos Antineoplásicos/administración & dosificación , Cardiotoxicidad/tratamiento farmacológico , Cardiotoxicidad/etiología , Enfermedades Cardiovasculares/enzimología , Ciclooxigenasa 2/metabolismo , Fluorouracilo/administración & dosificación , Masculino , Miocardio/enzimología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento , Factor de Necrosis Tumoral alfa/metabolismo
5.
Anal Biochem ; 639: 114508, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-34871563

RESUMEN

The lysyl oxidase (LOX) family of enzymes catalyze the oxidative deamination of lysine and hydroxylysine residues in collagen and elastin in the initiation step of the formation of covalent cross-linkages, an essential process for extracellular matrix (ECM) maturation. Elevated LOX expression levels leading to increased LOX activity is associated with diverse pathologies including fibrosis, cancer, and cardiovascular diseases. Different protocols have been so far established to detect and quantify LOX activity from tissue samples and cultured cells, all of them showing advantages and drawbacks. This review article presents a critical overview of the main features of currently available methods as well as introduces some recent technologies called to revolutionize our approach to LOX catalysis.


Asunto(s)
Pruebas de Enzimas/métodos , Proteína-Lisina 6-Oxidasa/metabolismo , Animales , Técnicas Biosensibles/instrumentación , Técnicas Biosensibles/métodos , Enfermedades Cardiovasculares/enzimología , Pruebas de Enzimas/instrumentación , Humanos , Neoplasias/enzimología , Imagen Óptica/instrumentación , Imagen Óptica/métodos , Proteína-Lisina 6-Oxidasa/análisis
6.
Biomed Res Int ; 2021: 5584452, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34235221

RESUMEN

OBJECTIVE: According to reports, liver enzymes might play a role in the incidence and development of cardiometabolic diseases such as metabolic syndrome (MetS), hypertension (HTN), and cardiovascular diseases (CVD). We conducted a study to investigate this hypothesis among the Iranian Kurdish population. METHODS: We analyzed data from the baseline phase of the Ravansar noncommunicable disease (RaNCD) cohort. The association between liver enzymes (ALT, AST, ALT/AST ratio, GGT, and ALP) with cardiometabolic disease risk factors was investigated by multiple linear regression. The odds ratio of cardiometabolic diseases in each quartile category of liver enzyme concentration was estimated using multivariable logistic regression. RESULTS: The mean age of participants was 47.3 ± 4.1 years (48.1 years in males and 51.8 years in females). In the adjusted model, all enzymes were positively associated with MetS, HTN, and CVD risk factors except for the ALT/AST ratio with SBP and DBP. In the adjusted model, subjects in the fourth quartile for GGT, ALT/AST ratio, ALT, ALP, and AST had 3.29-, 2.94-, 2.45-, 2.00-, and 1.19-fold increased risk for MetS compared with subjects in the first quartile. Increased levels of GGT and ALP were positively associated with the risk of HTN (ORs = 1.33, 95%CI = 1.03-1.71 for GGT; ORs = 1.32, 95%CI = -1.68 for ALP). An increased GGT level was significantly associated with CVD (ORs = 1.54, 95%CI = 1.03-1.68). Within the normal range quartile, ALT had a significant correlation with the incidence of MetS. CONCLUSION: According to the present study, the levels of liver enzymes could be considered for early diagnosis of MetS, HTN, and CVD.


Asunto(s)
Enfermedades Cardiovasculares/enzimología , Hígado/enzimología , Adulto , Anciano , Presión Sanguínea , Enfermedades Cardiovasculares/epidemiología , Femenino , Humanos , Hipertensión/enzimología , Hipertensión/epidemiología , Incidencia , Irán/epidemiología , Irán/etnología , Masculino , Síndrome Metabólico/enzimología , Síndrome Metabólico/epidemiología , Persona de Mediana Edad , Análisis Multivariante , Oportunidad Relativa , Prevalencia , Estudios Prospectivos , Factores de Riesgo , Fumar
7.
Signal Transduct Target Ther ; 6(1): 183, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-33994545

RESUMEN

CK2 is a constitutively active Ser/Thr protein kinase, which phosphorylates hundreds of substrates, controls several signaling pathways, and is implicated in a plethora of human diseases. Its best documented role is in cancer, where it regulates practically all malignant hallmarks. Other well-known functions of CK2 are in human infections; in particular, several viruses exploit host cell CK2 for their life cycle. Very recently, also SARS-CoV-2, the virus responsible for the COVID-19 pandemic, has been found to enhance CK2 activity and to induce the phosphorylation of several CK2 substrates (either viral and host proteins). CK2 is also considered an emerging target for neurological diseases, inflammation and autoimmune disorders, diverse ophthalmic pathologies, diabetes, and obesity. In addition, CK2 activity has been associated with cardiovascular diseases, as cardiac ischemia-reperfusion injury, atherosclerosis, and cardiac hypertrophy. The hypothesis of considering CK2 inhibition for cystic fibrosis therapies has been also entertained for many years. Moreover, psychiatric disorders and syndromes due to CK2 mutations have been recently identified. On these bases, CK2 is emerging as an increasingly attractive target in various fields of human medicine, with the advantage that several very specific and effective inhibitors are already available. Here, we review the literature on CK2 implication in different human pathologies and evaluate its potential as a pharmacological target in the light of the most recent findings.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19 , Enfermedades Cardiovasculares , Quinasa de la Caseína II , Fibrosis Quística , Oftalmopatías , Trastornos Mentales , Inhibidores de Proteínas Quinasas/uso terapéutico , SARS-CoV-2 , COVID-19/enzimología , COVID-19/genética , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/genética , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/genética , Quinasa de la Caseína II/metabolismo , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/enzimología , Fibrosis Quística/genética , Oftalmopatías/tratamiento farmacológico , Oftalmopatías/enzimología , Oftalmopatías/genética , Humanos , Trastornos Mentales/tratamiento farmacológico , Trastornos Mentales/enzimología , Trastornos Mentales/genética , Mutación , Fosforilación , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
8.
Biomed Pharmacother ; 139: 111610, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33957567

RESUMEN

Sphingomyelin (SM) can be converted into ceramide (Cer) by neutral sphingomyelinase (NSM) and acid sphingomyelinase (ASM). Cer is a second messenger of lipids and can regulate cell growth and apoptosis. Increasing evidence shows that NSM and ASM play key roles in many processes, such as apoptosis, immune function and inflammation. Therefore, NSM and ASM have broad prospects in clinical treatments, especially in cancer, cardiovascular diseases (such as atherosclerosis), nervous system diseases (such as Alzheimer's disease), respiratory diseases (such as chronic obstructive pulmonary disease) and the phenotype of dwarfisms in adolescents, playing a complex regulatory role. This review focuses on the physiological functions of NSM and ASM and summarizes their roles in certain diseases and their potential applications in therapy.


Asunto(s)
Esfingomielina Fosfodiesterasa/fisiología , Animales , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/terapia , Humanos , Neoplasias/enzimología , Neoplasias/terapia , Enfermedades del Sistema Nervioso/enzimología , Enfermedades del Sistema Nervioso/terapia , Enfermedades Respiratorias/enzimología , Enfermedades Respiratorias/terapia , Esfingomielina Fosfodiesterasa/química , Esfingomielina Fosfodiesterasa/clasificación
9.
Am J Chin Med ; 49(4): 843-862, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33827385

RESUMEN

Neuraminidase, also known as sialidase, is ubiquitous in animals and microorganisms. It is predominantly distributed in the cell membrane, cytoplasmic vesicles, and lysosomes. Neuraminidase generally recognizes the sialic acid glycosidic bonds at the ends of glycoproteins or glycolipids and enzymatically removes sialic acid. There are four types of neuraminidases, named as Neu1, Neu2, Neu3, and Neu4. Among them, Neu1 is the most abundant in mammals. Recent studies have revealed the involvement of Neu1 in several diseases, including cardiovascular diseases, diabetes, cancers, and neurological disorders. In this review, we center the attention to the role of Neu1 in cardiovascular diseases, including atherosclerosis, ischemic myocardial injury, cerebrovascular disease, congenital heart disease, and pulmonary embolism. We also summarize inhibitors from Chinese herbal medicines (CHMs) in inhibiting virus neuraminidase or human Neu1. Many Chinese herbs and Chinese herb preparations, such as Lonicerae Japonicae Flos, Scutellariae Radix, Yupingfeng San, and Huanglian Jiedu Decoction, have neuraminidase inhibitory activity. We hope to highlight the emerging role of Neu1 in humans and potentially titillate interest for further studies in this area.


Asunto(s)
Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/enzimología , Medicamentos Herbarios Chinos/farmacología , Neuraminidasa/efectos de los fármacos , Neuraminidasa/metabolismo , Medicamentos Herbarios Chinos/química , Humanos , Estructura Molecular
10.
FEBS J ; 288(24): 7162-7182, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33405316

RESUMEN

Matrix metalloproteinases (MMPs) are zinc-dependent endopeptidases that were first discovered as proteases, which target and cleave extracellular proteins. During the past 20 years, however, intracellular roles of MMPs were uncovered and research on this new aspect of their biology expanded. MMP-2 is the first of this protease family to be reported to play a crucial intracellular role where it cleaves several sarcomeric proteins inside cardiac myocytes during oxidative stress-induced injury. Beyond MMP-2, currently at least eleven other MMPs are known to function intracellularly including MMP-1, MMP-3, MMP-7, MMP-8, MMP-9, MMP-10, MMP-11, MMP-12, MMP-14, MMP-23 and MMP-26. These intracellular MMPs are localized to different compartments inside the cell including the cytosol, sarcomere, mitochondria, and the nucleus. Intracellular MMPs contribute to the pathogenesis of various diseases. Cardiovascular renal disorders, inflammation, and malignancy are some examples. They also exert antiviral and bactericidal effects. Interestingly, MMPs can act intracellularly through both protease-dependent and protease-independent mechanisms. In this review, we will highlight the intracellular mechanisms of MMPs activation, their numerous subcellular locales, substrates, and roles in different pathological conditions. We will also discuss the future direction of MMP research and the necessity to exploit the knowledge of their intracellular targets and actions for the design of targeted inhibitors.


Asunto(s)
Enfermedades Cardiovasculares/enzimología , Metaloproteinasas de la Matriz/metabolismo , Neoplasias/enzimología , Animales , Humanos
11.
Int J Biochem Cell Biol ; 131: 105904, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33359085

RESUMEN

Nitric Oxide is a very well known gaseous second messenger molecule and vasorelaxant agent involved in a variety of signaling in the body such as neurotransmission, ion channel modulation, and inflammation modulation. However, it's reversible covalent attachment to thiol groups of cysteine residues under nitrosative stress leading to aberrant protein S-nitrosylation (PSNO) has been reported in several pathological conditions in the body stemming from neurodegenerative diseases, cancer, cardiovascular system, and immune system disorders. In the cell, PSNOs are partly unstable and transit to a more stable disulfide state serving as an intermediate step towards disulfide formation thus eliciting the biological response. Scientists have identified several cellular thiol-dependent disulfide reductases that have the intrinsic capability to reverse the modification by reducing the stable disulfides formed in PSNOs and thereby rescue S-nitrosylation-induced altered proteins. The physiological roles of these major cellular ubiquitous S-denitrosylases and their probable implementations have not been fully explored. Gaining knowledge from current research and development this review provides a deeper insight into understanding the interplay and role of the major ubiquitous S-denitrosylases in maintaining cellular redox homeostasis. This review umbrellas the mechanism of Thioredoxin, TRP14, and Glutaredoxin systems and highlights their substrates specificities at different cellular conditions, physiological roles, and importance in diseased conditions that would allow researchers to investigate effective therapeutic interventions for nitrosative stress-related diseases and disorders.


Asunto(s)
Enfermedades Cardiovasculares/enzimología , Glutarredoxinas/metabolismo , Enfermedades del Sistema Inmune/enzimología , Neoplasias/enzimología , Enfermedades Neurodegenerativas/enzimología , Tiorredoxinas/metabolismo , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , Cisteína/metabolismo , Regulación de la Expresión Génica , Glutarredoxinas/genética , Humanos , Enfermedades del Sistema Inmune/genética , Enfermedades del Sistema Inmune/patología , Neoplasias/genética , Neoplasias/patología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Óxido Nítrico/metabolismo , Nitrosación , Estrés Nitrosativo/genética , Oxidación-Reducción , S-Nitrosotioles/metabolismo , Transducción de Señal , Especificidad por Sustrato , Tiorredoxinas/genética
12.
Mol Cell Endocrinol ; 529: 111119, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33309638

RESUMEN

The identification of an alternate extended form of angiotensin I composed of the first twelve amino acids at the N-terminal of angiotensinogen has generated new knowledge of the importance of noncanonical mechanisms for renin independent generation of angiotensins. The human sequence of the dodecapeptide angiotensin-(1-12) [N-Asp1-Arg2-Val3-Tyr4-Ile5-His6-Pro7-Phe8-His9-Leu10-Val1-Ile12-COOH] is an endogenous substrate that in the rat has been documented to be present in multiple organs including the heart, brain, kidney, gut, adrenal gland, and the bone marrow. Newer studies have confirmed the existence of Ang-(1-12) as an Ang II-forming substrate in the blood and heart of normal and diseased patients. Studies to-date document that angiotensin II generation from angiotensin-(1-12) does not require renin participation while chymase rather than angiotensin converting enzyme shows high catalytic activity in converting this tissue substrate into angiotensin II directly.


Asunto(s)
Angiotensina II/metabolismo , Angiotensina I/metabolismo , Angiotensinógeno/metabolismo , Quimasas/metabolismo , Fragmentos de Péptidos/metabolismo , Sistema Renina-Angiotensina/genética , Glándulas Suprarrenales/enzimología , Angiotensina I/genética , Angiotensina II/genética , Angiotensinógeno/genética , Animales , Biocatálisis , Médula Ósea/enzimología , Encéfalo/enzimología , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , Quimasas/genética , Expresión Génica , Humanos , Intestinos/enzimología , Riñón/enzimología , Miocardio/enzimología , Fragmentos de Péptidos/genética , Ratas
13.
Int J Mol Sci ; 21(22)2020 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-33203121

RESUMEN

The protein acetylation of either the α-amino groups of amino-terminal residues or of internal lysine or cysteine residues is one of the major posttranslational protein modifications that occur in the cell with repercussions at the protein as well as at the metabolome level. The lysine acetylation status is determined by the opposing activities of lysine acetyltransferases (KATs) and lysine deacetylases (KDACs), which add and remove acetyl groups from proteins, respectively. A special group of KDACs, named sirtuins, that require NAD+ as a substrate have received particular attention in recent years. They play critical roles in metabolism, and their abnormal activity has been implicated in several diseases. Conversely, the modulation of their activity has been associated with protection from age-related cardiovascular and metabolic diseases and with increased longevity. The benefits of either activating or inhibiting these enzymes have turned sirtuins into attractive therapeutic targets, and considerable effort has been directed toward developing specific sirtuin modulators. This review summarizes the protein acylation/deacylation processes with a special focus on the current developments in the sirtuin research field.


Asunto(s)
Envejecimiento/metabolismo , Enfermedades Cardiovasculares/enzimología , Enfermedades Metabólicas/enzimología , Procesamiento Proteico-Postraduccional , Sirtuinas/metabolismo , Acetilación , Humanos
14.
Cardiovasc Toxicol ; 20(5): 448-453, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32632849

RESUMEN

Peroxiredoxin (Prx) refers to a family of thiol-dependent peroxidases that decompose hydrogen peroxide, lipid hydroperoxides, as well as peroxynitrite, and protect against oxidative and inflammatory stress. There are six mammalian Prx isozymes (Prx1-6), classified as typical 2-Cys, atypical 2-Cys, or 1-Cys Prxs based on the mechanism and the number of cysteine residues involved during catalysis. In addition to their well-established peroxide-scavenging activity, some Prxs also participate in the regulation of various cell signaling pathways. Extensive animal studies employing primarily gene knockout models provide substantial evidence supporting a critical protective role of Prxs in various disease processes involving oxidative and inflammatory stress. This review surveys recent research findings, published primarily in influential journals, on the involvement of various Prx isozymes in protecting against cardiovascular injury and related disorders, including diabetes, metabolic syndromes, and sepsis, whose pathophysiology all intimately involves oxidative stress and inflammation.


Asunto(s)
Enfermedades Cardiovasculares/prevención & control , Miocitos Cardíacos/enzimología , Peroxirredoxinas/metabolismo , Animales , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/fisiopatología , Humanos , Mediadores de Inflamación/metabolismo , Isoenzimas , Miocitos Cardíacos/patología , Estrés Oxidativo , Peroxirredoxinas/genética , Transducción de Señal
15.
Int J Mol Sci ; 21(15)2020 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-32722521

RESUMEN

Arginase is a widely known enzyme of the urea cycle that catalyzes the hydrolysis of L-arginine to L-ornithine and urea. The action of arginase goes beyond the boundaries of hepatic ureogenic function, being widespread through most tissues. Two arginase isoforms coexist, the type I (Arg1) predominantly expressed in the liver and the type II (Arg2) expressed throughout extrahepatic tissues. By producing L-ornithine while competing with nitric oxide synthase (NOS) for the same substrate (L-arginine), arginase can influence the endogenous levels of polyamines, proline, and NO•. Several pathophysiological processes may deregulate arginase/NOS balance, disturbing the homeostasis and functionality of the organism. Upregulated arginase expression is associated with several pathological processes that can range from cardiovascular, immune-mediated, and tumorigenic conditions to neurodegenerative disorders. Thus, arginase is a potential biomarker of disease progression and severity and has recently been the subject of research studies regarding the therapeutic efficacy of arginase inhibitors. This review gives a comprehensive overview of the pathophysiological role of arginase and the current state of development of arginase inhibitors, discussing the potential of arginase as a molecular imaging biomarker and stimulating the development of novel specific and high-affinity arginase imaging probes.


Asunto(s)
Arginasa/metabolismo , Biomarcadores de Tumor/metabolismo , Enfermedades Cardiovasculares/enzimología , Enfermedades del Sistema Inmune/enzimología , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimología , Enfermedades Neurodegenerativas/enzimología , Animales , Humanos , Óxido Nítrico Sintasa/metabolismo
16.
J Cell Mol Med ; 24(18): 10274-10278, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32713161

RESUMEN

With the outbreak of a new coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the public healthcare systems are facing great challenges. Coronavirus disease 2019 (COVID-19) could develop into severe pneumonia, acute respiratory distress syndrome and multi-organ failure. Remarkably, in addition to the respiratory symptoms, some COVID-19 patients also suffer from cardiovascular injuries. Dipeptidyl peptidase-4 (DPP-4) is a ubiquitous glycoprotein which could act both as a cell membrane-bound protein and a soluble enzymatic protein after cleavage and release into the circulation. Despite angiotensin-converting enzyme 2 (ACE2), the recently recognized receptor of SARS-CoV and SARS-CoV-2, which facilitated their entries into the host, DPP-4 has been identified as the receptor of middle east respiratory syndrome coronavirus (MERS-CoV). In the current review, we discussed the potential roles of DPP-4 in COVID-19 and the possible effects of DPP-4 inhibitors on cardiovascular system in patients with COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , Enfermedades Cardiovasculares/enzimología , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Enfermedades Cardiovasculares/virología , Interacciones Huésped-Patógeno , Humanos , SARS-CoV-2/fisiología , Internalización del Virus
17.
Cell Mol Biol (Noisy-le-grand) ; 66(3): 57-64, 2020 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-32538748

RESUMEN

Cardiovascular disease (CVDs) is the leading cause of morbidity and death worldwide. Most genetic variants could be identified by several genome-wide-association-studies (GWAS), including within genes encoding proteins involved in the AKT/PI3K pathways that are related with an increased risk of metabolic syndrome and CVDs. Therefore, due to the importance of genetic variants in the prognosis of diseases, we examined the genetic polymorphism of AKT-rs1130233 located on chromosome 14 with cardiovascular risk factors. In this cross-sectional study, 721 subjects recruited from the Mashhad-Stroke and Heart-Atherosclerotic-Disorders (MASHAD) cohort study. The participants including 257 subjects with metabolic syndrome, 144 subjects with cardiovascular disease and 320 subjects as a control group. Anthropometric, biochemical and demographic information measures were prepared. Dietary assessment was managed by 24h dietary recall. DNA extraction and genotyping were carried out by using the TaqMan real-time-PCR based method. The association of AKT rs1130233 locus with dietary intakes, metabolic syndrome and cardiovascular risk factors were assessed. Data were analyzed by using SPSS 21 software. Frequencies of genotypes AA, AG and GG of the AKT rs1130233 polymorphism were 12.6%, 44.5% and 42.9% in subjects with metabolic syndrome and 9.7%, 39.6% and 50.7% in subjects with cardiovascular disease, respectively. The frequency of allele A and G in cardiovascular disease and metabolic syndrome population were 29.5%, 70.5% and 34.8%, 65.2%, respectively. We have found no significant association between the AKT rs1130233 polymorphism with cardiovascular risk factors and metabolic syndrome. The results of dietary intake showed that the levels of phosphorus intake (p=0.008), calcium intake (p=0.007) and iodine intake (p=0.04) were different in subjects with and without metabolic syndrome. And also, energy intake was significantly different in subjects with cardiovascular disease (p=0.01) compared to the control group. Our findings suggest that AKT rs1130233 was not associated with the risk of metabolic syndrome and cardiovascular disease in the Iranian population. More studies are needed to validate our results. We did functional analysis, due to certify our investigation about value of this genetic biomarker for CVD risk.


Asunto(s)
Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/genética , Sitios Genéticos , Predisposición Genética a la Enfermedad , Variación Genética , Estudio de Asociación del Genoma Completo , Proteínas Proto-Oncogénicas c-akt/genética , Adulto , Dieta , Femenino , Frecuencia de los Genes/genética , Humanos , Masculino , Síndrome Metabólico/genética , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Factores de Riesgo
18.
Pharmacol Res ; 159: 104990, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32505836

RESUMEN

Sestrin2 is a cysteine sulfinyl reductase that plays crucial roles in regulation of antioxidant actions. Sestrin2 provides cytoprotection against multiple stress conditions, including hypoxia, endoplasmic reticulum (ER) stress and oxidative stress. Recent research reveals that upregulation of Sestrin2 is induced by various transcription factors such as p53 and activator protein 1 (AP-1), which further promotes AMP-activated protein kinase (AMPK) activation and inhibits mammalian target of rapamycin protein kinase (mTOR) signaling. Sestrin2 triggers autophagy activity to reduce cellular reactive oxygen species (ROS) levels by promoting nuclear factor erythroid 2 (NF-E2)-related factor 2 (Nrf2) activation and Kelch-like ECH-associated protein 1 (Keap1) degradation, which plays a pivotal role in homeostasis of metabolic regulation. Under hypoxia and ER stress conditions, elevated Sestrin2 expression maintains cellular homeostasis through regulation of antioxidant genes. Sestrin2 is responsible for diminishing cellular ROS accumulation through autophagy via AMPK activation, which displays cardioprotection effect in cardiovascular diseases. In this review, we summarize the recent understanding of molecular structure, biological roles and biochemical functions of Sestrin2, and discuss the roles and mechanisms of Sestrin2 in autophagy, hypoxia and ER stress. Understanding the precise functions and exact mechanism of Sestrin2 in cellular homeostasis will provide the evidence for future experimental research and aid in the development of novel therapeutic strategies for cardiovascular diseases.


Asunto(s)
Autofagia , Enfermedades Cardiovasculares/enzimología , Sistema Cardiovascular/enzimología , Proteínas Nucleares/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Autofagia/efectos de los fármacos , Fármacos Cardiovasculares/uso terapéutico , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/patología , Sistema Cardiovascular/efectos de los fármacos , Sistema Cardiovascular/patología , Hipoxia de la Célula , Humanos , Terapia Molecular Dirigida , Factor 2 Relacionado con NF-E2/metabolismo , Proteínas Nucleares/efectos de los fármacos , Estrés Oxidativo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
19.
Arthritis Rheumatol ; 72(10): 1658-1667, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32475073

RESUMEN

OBJECTIVE: To investigate the relationship between anti-citrullinated protein antibodies (ACPAs), specific ACPA subspecificities, rheumatoid factor (RF) isotypes, and incident cardiovascular (CV) events in patients with rheumatoid arthritis (RA). METHODS: Serum samples from Swedish patients with new-onset RA (diagnosed within 1 year of symptom onset between 1996 and 2009) were centrally typed for anti-cyclic citrullinated peptide 2 (anti-CCP2) antibodies, 20 ACPA subspecificities, and RF isotypes. Patients were followed up longitudinally in nationwide registers to monitor the occurrence of acute coronary syndrome (ACS), stroke, CV-related death, and major adverse CV events (MACE). The association between each serologic marker and CV outcome, and the impact of adjustment for the Disease Activity Score in 28 joints (DAS28), smoking status, and income at baseline, were assessed using Cox proportional hazards models. In addition, associations of serologic markers with all-cause mortality were explored. RESULTS: In total, 2,814 patients with RA were included in the study. The median follow-up was 13 years, during which the CV end points of ACS, stroke, or CV-related death were reported to occur in 375 patients. Occurrence and/or levels of anti-CCP2 were associated with risk of incident ACS (hazard ratio [HR] 1.46, 95% confidence interval [95% CI] 1.03-2.06), stroke (HR 1.47, 95% CI 1.03-2.10), CV-related death (P = 0.024 for association with anti-CCP2 levels), and MACE (HR 1.34, 95% CI 1.06-1.70). Similarly, an association with the number of ACPA subspecificities was observed; however, this could not be attributed to any individual or group of ACPA subspecificities. Presence of IgM-RF was associated with all CV end points except ACS, and IgA-RF was exclusively associated with CV-related death. Adjustment for smoking status, income, and DAS28 scores decreased most of the HRs, whereas IgA-RF remained associated with CV-related death (HR 1.61, 95% CI 1.05-2.48). All of the assessed serologic makers were associated with all-cause mortality. CONCLUSION: RF isotypes and ACPAs are associated with future CV events in patients with RA. ACPA levels and number of subspecificities seem more important than the occurrence of particular subspecificities, and these associations were not explained by a history of ever smoking.


Asunto(s)
Artritis Reumatoide/inmunología , Enfermedades Cardiovasculares/enzimología , Isotipos de Inmunoglobulinas/sangre , Factor Reumatoide/inmunología , Adulto , Anciano , Anticuerpos Antiproteína Citrulinada/sangre , Especificidad de Anticuerpos , Artritis Reumatoide/sangre , Artritis Reumatoide/complicaciones , Enfermedades Cardiovasculares/etiología , Estudios de Casos y Controles , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Suecia
20.
Biomed Pharmacother ; 129: 110354, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32540644

RESUMEN

ALOX12 encodes arachidonic acid 12-lipoxygenase that acts on different polyunsaturated fatty acid substrates to produce biologically active lipid mediators including eicosanes and lipoxins. ALOX12 protein plays an important role in inflammation and oxidation, while abnormal DNA methylation and genetic variants of ALOX12 are associated with various human diseases and pathological phenotypes, such as cardiovascular disease, diabetes, neurodegenerative diseases, respiratory system disease, cancer, infection, etc. Here, this article reviews the mechanisms by which ALOX12 participates in related diseases, which will provide systematic knowledge for future ALOX12 related studies.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Animales , Araquidonato 12-Lipooxigenasa/genética , Enfermedades Cardiovasculares/enzimología , Enfermedades Cardiovasculares/genética , Predisposición Genética a la Enfermedad , Humanos , Inflamación/enzimología , Inflamación/genética , Enfermedades Metabólicas/enzimología , Enfermedades Metabólicas/genética , Neoplasias/enzimología , Neoplasias/genética , Enfermedades del Sistema Nervioso/enzimología , Enfermedades del Sistema Nervioso/genética , Fenotipo , Polimorfismo de Nucleótido Simple , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA