Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Itamar Couto Guedes de Jesus is active.

Publication


Featured researches published by Itamar Couto Guedes de Jesus.


Frontiers in Physiology | 2017

Vascular Kinin B1 and B2 Receptors Determine Endothelial Dysfunction through Neuronal Nitric Oxide Synthase

Thássio Ricardo Ribeiro Mesquita; Gianne P. Campos-Mota; Virginia S. Lemos; Jader Santos Cruz; Itamar Couto Guedes de Jesus; Enilton A. Camargo; Jorge L. Pesquero; João Bosco Pesquero; Luciano S. A. Capettini; Sandra Lauton-Santos

B1- and B2-kinin receptors are G protein-coupled receptors that play an important role in the vascular function. Therefore, the present study was designed to evaluate the participation of kinin receptors in the acetylcholine (ACh)-induced vascular relaxation, focusing on the protein-protein interaction involving kinin receptors with endothelial and neuronal nitric oxide synthases (eNOS and nNOS). Vascular reactivity, nitric oxide (NO·) and reactive oxygen species (ROS) generation, co-immunoprecipitation were assessed in thoracic aorta from male wild-type (WT), B1- (B1R−/−), B2- (B2R−/−) knockout mice. Some vascular reactivity experiments were also performed in a double kinin receptors knockout mice (B1B2R−/−). For pharmacological studies, selective B1- and B2-kinin receptors antagonists, NOS inhibitors and superoxide dismutase (SOD) mimetic were used. First, we show that B1- and B2-kinin receptors form heteromers with nNOS and eNOS in thoracic aorta. To investigate the functionality of these protein-protein interactions, we took advantage of pharmacological tools and knockout mice. Importantly, our results show that kinin receptors regulate ACh-induced relaxation via nNOS signaling in thoracic aorta with no changes in NO· donor-induced relaxation. Interestingly, B1B2R−/− presented similar level of vascular dysfunction as found in B1R−/− or B2R−/− mice. In accordance, aortic rings from B1R−/− or B2R−/− mice exhibit decreased NO· bioavailability and increased superoxide generation compared to WT mice, suggesting the involvement of excessive ROS generation in the endothelial dysfunction of B1R−/− and B2R−/− mice. Alongside, we show that impaired endothelial vasorelaxation induced by ACh in B1R−/− or B2R−/− mice was rescued by the SOD mimetic compound. Taken together, our findings show that B1- and B2-kinin receptors regulate the endothelium-dependent vasodilation of ACh through nNOS activity and indicate that molecular disturbance of short-range interaction between B1- and B2-kinin receptors with nNOS might be involved in the oxidative pathogenesis of endothelial dysfunction.


Frontiers in Pharmacology | 2017

Cardioprotective Action of Ginkgo biloba Extract against Sustained β-Adrenergic Stimulation Occurs via Activation of M2/NO Pathway

Thássio Ricardo Ribeiro Mesquita; Itamar Couto Guedes de Jesus; Jucilene F. dos Santos; Grace Kelly Melo de Almeida; Carla Maria Lins de Vasconcelos; Silvia Guatimosim; Fabricio Macedo; Robervan Santos; José Evaldo Rodrigues de Menezes-Filho; Rodrigo Miguel-dos-Santos; Paulo Tojal Dantas Matos; Sergio Scalzo; Valter J. Santana-Filho; Ricardo Luiz Cavalcanti De Albuquerque-Júnior; Rose Nely Pereira-Filho; Sandra Lauton-Santos

Ginkgo biloba is the most popular phytotherapic agent used worldwide for treatment of several human disorders. However, the mechanisms involved in the protective actions of Ginkgo biloba on cardiovascular diseases remain poorly elucidated. Taking into account recent studies showing beneficial actions of cholinergic signaling in the heart and the cholinergic hypothesis of Ginkgo biloba-mediated neuroprotection, we aimed to investigate whether Ginkgo biloba extract (GBE) promotes cardioprotection via activation of cholinergic signaling in a model of isoproterenol-induced cardiac hypertrophy. Here, we show that GBE treatment (100 mg/kg/day for 8 days, v.o.) reestablished the autonomic imbalance and baroreflex dysfunction caused by chronic β-adrenergic receptor stimulation (β-AR, 4.5 mg/kg/day for 8 days, i.p.). Moreover, GBE prevented the upregulation of muscarinic receptors (M2) and downregulation of β1-AR in isoproterenol treated-hearts. Additionally, we demonstrated that GBE prevents the impaired endothelial nitric oxide synthase activity in the heart. GBE also prevented the pathological cardiac remodeling, electrocardiographic changes and impaired left ventricular contractility that are typical of cardiac hypertrophy. To further investigate the mechanisms involved in GBE cardioprotection in vivo, we performed in vitro studies. By using neonatal cardiomyocyte culture we demonstrated that the antihypertrophic action of GBE was fully abolished by muscarinic receptor antagonist or NOS inhibition. Altogether, our data support the notion that antihypertrophic effect of GBE occurs via activation of M2/NO pathway uncovering a new mechanism involved in the cardioprotective action of Ginkgo biloba.


Basic & Clinical Pharmacology & Toxicology | 2017

Dissection of the Effects of Quercetin on Mouse Myocardium

Michel Santana Santos; Evaleide Diniz de Oliveira; Artur Santos-Miranda; Jader Santos Cruz; Antônio Nei Santana Gondim; José Evaldo Rodrigues de Menezes-Filho; Diego Santos Souza; Leidiane Pinho-da-Silva; Itamar Couto Guedes de Jesus; Danilo Roman-Campos; Silvia Guatimosim; Aline Lara; Eduardo Antonio Conde-Garcia; Carla Maria Lins de Vasconcelos

Quercetin is a plant flavonoid with several biological activities. This study aimed to describe quercetin effects on contractile and electrophysiological properties of the cardiac muscle as well as on calcium handling. Quercetin elicited positive inotropism that was significantly reduced by propranolol indicating an involvement of the sympathetic nervous system. In cardiomyocytes, 30 μM quercetin increased ICa,L at 0 mV from −0.95 ± 0.01 A/F to −1.21 ± 0.08 A/F. The membrane potential at which 50% of the channels are activated (V0.5) shifted towards more negative potentials from −13.06 ± 1.52 mV to −19.26 ± 1.72 mV and did not alter the slope factor. Furthermore, quercetin increased [Ca2+]i transient by 28% when compared to control. Quercetin accelerated [Ca2+]i transient decay time, which could be attributed to SERCA activation. In resting cardiomyocytes, quercetin did not change amplitude or frequency of Ca2+ sparks. In isolated heart, quercetin increased heart rate and decreased PRi, QTc and duration of the QRS complex. Thus, we showed that quercetin activates β‐adrenoceptors, leading to increased L‐type Ca2+ current and cell‐wide intracellular Ca2+ transient without visible changes in Ca2+ sparks.


Neurochemistry International | 2018

Neuromuscular synapse degeneration without muscle function loss in the diaphragm of a murine model for Huntington's Disease

Priscila Aparecida Costa Valadão; Matheus P.S.M. Gomes; Bárbara Campos de Aragão; Hermann A. Rodrigues; Jéssica N. Andrade; Rubens Garcias; Julliane V. Joviano-Santos; Murilo A. Luiz; Wallace L. Camargo; Lígia Araujo Naves; Christopher Kushmerick; Walter L.G. Cavalcante; Márcia Gallacci; Itamar Couto Guedes de Jesus; Silvia Guatimosim; Cristina Guatimosim

ABSTRACT Huntingtons disease (HD) is an autosomal dominant neurodegenerative disease characterized by chorea, incoordination and psychiatric and behavioral symptoms. The leading cause of death in HD patients is aspiration pneumonia, associated with respiratory dysfunction, decreased respiratory muscle strength and dysphagia. Although most of the motor symptoms are derived from alterations in the central nervous system, some might be associated with changes in the components of motor units (MU). To explore this hypothesis, we evaluated morphofunctional aspects of the diaphragm muscle in a mouse model for HD (BACHD). We showed that the axons of the phrenic nerves were not affected in 12‐months‐old BACHD mice, but the axon terminals that form the neuromuscular junctions (NMJs) were more fragmented in these animals in comparison with the wild‐type mice. In BACHD mice, the synaptic vesicles of the diaphragm NMJs presented a decreased exocytosis rate. Quantal content and quantal size were smaller and there was less synaptic depression whereas the estimated size of the readily releasable vesicle pool was not changed. At the ultrastructure level, the diaphragm NMJs of these mice presented fewer synaptic vesicles with flattened and oval shapes, which might be associated with the reduced expression of the vesicular acetylcholine transporter protein. Furthermore, mitochondria of the diaphragm muscle presented signs of degeneration in BACHD mice. Interestingly, despite all these cellular alterations, BACHD diaphragmatic function was not compromised, suggesting a higher resistance threshold of this muscle. A putative resistance mechanism may be protecting this vital muscle. Our data contribute to expanding the current understanding of the effects of mutated huntingtin in the neuromuscular synapse and the diaphragm muscle function. HIGHLIGHTSAxons from the phrenic nerve were not affected in 12‐months‐old BACHD mice but axon terminals (NMJs) were more fragmented.Synaptic vesicles from the NMJs of BACHD presented decreased exocytosis rate, abnormal shape and released less acetylcholine.Diaphragm muscle mitochondria presented signs of degeneration in BACHD altough muscle function was preserved.


Journal of Molecular and Cellular Cardiology | 2018

Resistance exercise mediates remote ischemic preconditioning by limiting cardiac eNOS uncoupling

Michael Nadson Santos Santana; Diego Santos Souza; Rodrigo Miguel-dos-Santos; Thallita Kelly Rabelo; Carla Maria Lins de Vasconcelos; Juliana Maria Navia-Pelaez; Itamar Couto Guedes de Jesus; Júlio Alves da Silva-Neto; Sandra Lauton-Santos; Luciano S. A. Capettini; Silvia Guatimosim; Russell G. Rogers; Márcio R. V. Santos; Valter J. Santana-Filho; Thássio Ricardo Ribeiro Mesquita

BACKGROUND Currently viewed as a complementary non-pharmacological intervention for preventing cardiac disorders, long-term aerobic training produces cardioprotection through remote ischemic preconditioning (RIPC) mechanisms. However, RIPC triggered by acute exercise remains poorly understood. Although resistance exercise (RE) has been highly recommended by several public health guidelines, there is no evidence showing that RE mediates RIPC. Hence, we investigated whether RE induces cardiac RIPC through nitric oxide synthase (NOS)-dependent mechanism. METHODS AND RESULTS Acute RE at 40% of the maximal load augmented systemic nitrite levels, associated with increased cardiac eNOS phosphorylation, without affecting nNOS activity. Using an experimental model of myocardial infarction (MI) through ischemia-reperfusion (IR), RE fully prevented the loss of cardiac contractility and the extent of MI size compared to non-exercised (NE) rats. Moreover, RE mitigated aberrant ST-segment and reduced life-threatening arrhythmias induced by IR. Importantly, inhibition of NOS abolished the RE-mediated cardioprotection. After IR, NE rats showed increased cardiac eNOS activity, associated with reduced dimer/monomer ratio. Supporting the pivotal role of eNOS coupling during MI, non-exercised rats displayed a marked generation of reactive oxygen species (ROS) and oxidative-induced carbonylation of proteins, whereas RE prevented these responses. We validated our data demonstrating a restoration of physiological ROS levels in NE + IR cardiac sections treated with BH4, a cofactor oxidatively depleted during eNOS uncoupling, while cardiac ROS generation from exercised rats remained unchanged, suggesting no physiological needs of supplemental eNOS cofactors. CONCLUSION Together, our findings strongly indicate that RE mediates RIPC by limiting eNOS uncoupling and mitigates myocardial IR injury.


Free Radical Biology and Medicine | 2018

Endurance training restores spatially distinct cardiac mitochondrial function and myocardial contractility in ovariectomized rats

Elis Aguiar Morra; Paula Lopes Rodrigues; Itamar Couto Guedes de Jesus; Patrícia Ribeiro Do Val Lima; Renata Andrade Ávila; Tadeu Ériton Caliman Zanardo; Breno Valentim Nogueira; Donald M. Bers; Silvia Guatimosim; Ivanita Stefanon; Rogério Faustino Ribeiro Júnior

ABSTRACT We previously demonstrated that the loss of female hormones induces cardiac and mitochondrial dysfunction in the female heart. Here, we show the impact of endurance training for twelve weeks, a nonpharmacological therapy against cardiovascular disease caused by ovariectomy and its contribution to cardiac contractility, mitochondrial quality control, bioenergetics and oxidative damage. We found that ovariectomy induced cardiac hypertrophy and dysfunction by decreasing SERCA2 and increasing phospholamban protein expression. Endurance training restored myocardial contractility, SERCA2 levels, increased calcium transient in ovariectomized rats but did not change phospholamban protein expression or cardiac hypertrophy. Additionally, ovariectomy decreased the amount of intermyofibrillar mitochondria and induced mitochondrial fragmentation that were accompanied by decreased levels of mitofusin 1, PGC‐1&agr;, NRF‐1, total AMPK‐&agr; and mitochondrial Tfam. Endurance training prevented all these features except for mitofusin 1. Ovariectomy reduced O2 consumption, elevated O2.‐ release and increased Ca2+‐induced mitochondrial permeability transition pore opening in both mitochondrial subpopulations. Ovariectomy also increased NOX‐4 protein expression in the heart, reduced mitochondrial Mn‐SOD, catalase protein expression and increased protein carbonylation in both mitochondrial subpopulations, which were prevented by endurance training. Taken together, our findings show that endurance training prevented cardiac contractile dysfunction and mitochondrial quality control in ovariectomized rats. Graphical abstract Figure. No Caption available.


American Journal of Physiology-cell Physiology | 2017

Absence of suppressor of cytokine signaling 2 turns cardiomyocytes unresponsive to LIF-dependent increases in Ca2+ levels

Cibele Rocha-Resende; Itamar Couto Guedes de Jesus; Danilo Roman-Campos; Artur S. Miranda; Fabiana Alves; Rodrigo R Resende; Jader Santos Cruz; Fabiana S. Machado; Silvia Guatimosim

Little is known regarding the role of suppressor of cytokine signaling (SOCS) in the control of cytokine signaling in cardiomyocytes. We investigated the consequences of SOCS2 ablation for leukemia inhibitory factor (LIF)-induced enhancement of intracellular Ca2+ ([Ca2+]i) transient by performing experiments with cardiomyocytes from SOCS2-knockout (ko) mice. Similar levels of SOCS3 transcripts were seen in cardiomyocytes from wild-type and SOCS2-ko mice, while SOCS1 mRNA was reduced in SOCS2-ko. Immunoprecipitation experiments showed increased SOCS3 association with gp130 receptor in SOCS2-ko myocytes. Measurements of Ca2+ in wild-type myocytes exposed to LIF showed a significant increase in the magnitude of the Ca2+ transient. This change was absent in LIF-treated SOCS2-ko cells. LIF activation of ERK and STAT3 was observed in both wild-type and SOCS2-ko cells, indicating that in SOCS2-ko, LIF receptors were functional, despite the lack of effect in the Ca2+ transient. In wild-type cells, LIF-induced increase in [Ca2+]i and phospholamban Thr17 [PLN(Thr17)] phosphorylation was inhibited by KN-93, indicating a role for CaMKII in LIF-induced Ca2+ raise. LIF-induced phosphorylation of PLN(Thr17) was abrogated in SOCS2-ko myocytes. In wild-type cardiomyocytes, LIF treatment increased L-type Ca2+ current (ICa,L), a key activator of CaMKII in response to LIF. Conversely, SOCS2-ko myocytes failed to activate ICa,L in response to LIF, providing a rationale for the lack of LIF effect on Ca2+ transient. Our data show that absence of SOCS2 turns cardiomyocytes unresponsive to LIF-induced [Ca2+] raise, indicating that endogenous levels of SOCS2 are crucial for full activation of LIF signaling in the heart.


Neurochemistry International | 2018

Corrigendum to “Neuromuscular synapse degeneration without muscle function loss in the diaphragm of a murine model for Huntington's Disease” [Neurochem. Int. 116 (2018) 30–42]

Priscila Aparecida Costa Valadão; Matheus P.S.M. Gomes; Bárbara Campos de Aragão; Hermann Alecsandro Rodrigues; Jéssica N. Andrade; Rubens Garcias; Julliane V. Joviano-Santos; Murilo A. Luiz; Wallace L. Camargo; Lígia Araujo Naves; Christopher Kushmerick; Walter L.G. Cavalcante; Márcia Gallacci; Itamar Couto Guedes de Jesus; Silvia Guatimosim; Cristina Guatimosim


Molecular and Cellular Endocrinology | 2018

Testosterone deficiency prevents left ventricular contractility dysfunction after myocardial infarction

R.F. Ribeiro Júnior; Karoline de Sousa Ronconi; Itamar Couto Guedes de Jesus; P.W.M. Almeida; L. Forechi; Dalton Valentim Vassallo; Silvia Guatimosim; Ivanita Stefanon; Aurélia Araújo Fernandes


Food and Chemical Toxicology | 2018

Myrtenol protects against myocardial ischemia-reperfusion injury through antioxidant and anti-apoptotic dependent mechanisms

Raquel Moreira de Britto; Júlio Alves da Silva-Neto; Thássio Ricardo Ribeiro Mesquita; Carla Maria Lins de Vasconcelos; Grace Kelly Melo de Almeida; Itamar Couto Guedes de Jesus; Péligris Henrique dos Santos; Diego Santos Souza; Rodrigo Miguel-dos-Santos; Lucas Andrade de Sá; Fanildes Silva Moraes dos Santos; Rose Nely Pereira-Filho; Ricardo Luiz Cavalcanti De Albuquerque-Júnior; Lucindo J. Quintans-Júnior; Silvia Guatimosim; Sandra Lauton-Santos

Collaboration


Dive into the Itamar Couto Guedes de Jesus's collaboration.

Top Co-Authors

Avatar

Silvia Guatimosim

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sandra Lauton-Santos

Universidade Federal de Sergipe

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Diego Santos Souza

Universidade Federal de Sergipe

View shared research outputs
Top Co-Authors

Avatar

Jader Santos Cruz

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bárbara Campos de Aragão

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar

Christopher Kushmerick

Universidade Federal de Minas Gerais

View shared research outputs
Top Co-Authors

Avatar

Cibele Rocha-Resende

Universidade Federal de Minas Gerais

View shared research outputs
Researchain Logo
Decentralizing Knowledge