Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Curr Pharm Des ; 23(40): 6132-6141, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28847304

RESUMO

Preterm birth (PTB) is a leading cause of neonatal mortality and morbidity worldwide, and surviving infants are at increased risks of lifelong complications. PTB has been firmly linked to inflammation regardless of infection, specific aetiology or timing of birth. Deleterious inflammation is observed in maternal and fetal tissue, and correlates with the severity of perinatal complications. At present, PTB is treated with tocolytics as though it is exclusively a myometrial contractile disorder. These agents do not address underlying inflammatory processes and are thus vastly ineffective at improving neonatal outcomes. Of all inflammatory mediators, IL-1 is central to the pathophysiology of PTB and most adverse neonatal outcomes. We thus present herein a review of the various effects of IL-1 in utero, with a brief overview of its mechanism of action. We then discuss the potential of different IL-1-targeting agents based on pre-clinical testing in relevant models of PTB and neonatal inflammatory injuries.


Assuntos
Inflamação/tratamento farmacológico , Interleucina-1/antagonistas & inibidores , Nascimento Prematuro , Receptores de Interleucina-1/antagonistas & inibidores , Animais , Feminino , Inflamação/metabolismo , Inflamação/patologia , Interleucina-1/metabolismo , Gravidez , Receptores de Interleucina-1/metabolismo
2.
Neurology ; 88(6): 551-561, 2017 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-28087824

RESUMO

OBJECTIVE: To seek a neuropathologic signature of sudden unexpected death in epilepsy (SUDEP) in a postmortem cohort by use of immunohistochemistry for specific markers of inflammation, gliosis, acute neuronal injury due to hypoxia, and blood-brain barrier (BBB) disruption, enabling the generation of hypotheses about potential mechanisms of death in SUDEP. METHODS: Using immunohistochemistry, we investigated the expression of 6 markers (CD163, human leukocyte antigen-antigen D related, glial fibrillary acid protein, hypoxia-inducible factor-1α [HIF-1α], immunoglobulin G, and albumin) in the hippocampus, amygdala, and medulla in 58 postmortem cases: 28 SUDEP (definite and probable), 12 epilepsy controls, and 18 nonepileptic sudden death controls. A semiquantitative measure of immunoreactivity was scored for all markers used, and quantitative image analysis was carried out for selected markers. RESULTS: Immunoreactivity was observed for all markers used within all studied brain regions and groups. Immunoreactivity for inflammatory reaction, BBB leakage, and HIF-1α in SUDEP cases was not different from that seen in control groups. CONCLUSIONS: This study represents a starting point to explore by immunohistochemistry the mechanisms underlying SUDEP in human brain tissue. Our approach highlights the potential and importance of considering immunohistochemical analysis to help identify biomarkers of SUDEP. Our results suggest that with the markers used, there is no clear immunohistochemical signature of SUDEP in human brain.


Assuntos
Encéfalo/metabolismo , Encéfalo/patologia , Morte Súbita , Epilepsia/metabolismo , Epilepsia/patologia , Adolescente , Adulto , Biomarcadores/metabolismo , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Encéfalo/irrigação sanguínea , Feminino , Humanos , Hipóxia/metabolismo , Hipóxia/patologia , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neuroglia/metabolismo , Neuroglia/patologia , Neuroimunomodulação , Neurônios/metabolismo , Neurônios/patologia , Adulto Jovem
3.
Am J Obstet Gynecol ; 216(1): 60.e1-60.e17, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27615440

RESUMO

BACKGROUND: Uterine inflammatory processes trigger prolabor pathways and orchestrate on-time labor onset. Although essential for successful labor, inflammation needs to be regulated to avoid uncontrolled amplification and resolve postpartum. During labor, myometrial smooth muscle cells generate ATP mainly via anaerobic glycolysis, resulting in accumulation of lactate. Aside from its metabolic function, lactate has been shown to activate a G protein-coupled receptor, GPR81, reported to regulate inflammation. We therefore hypothesize that lactate produced during labor may act via GPR81 in the uterus to exert in a feedback manner antiinflammatory effects, to resolve or mitigate inflammation. OBJECTIVE: We sought to investigate the role of lactate produced during labor and its receptor, GPR81, in regulating inflammation in the uterus. STUDY DESIGN: We investigated the expression of GPR81 in the uterus and the pharmacological role of lactate acting via GPR81 during labor, using shRNA-GPR81 and GPR81-/- mice. RESULTS: (1) Uterine lactate levels increased substantially from 2 to 9 mmol/L during labor. (2) Immunohistological analysis revealed expression of GPR81 in the uterus with high expression in myometrium. (3) GPR81 expression increased during gestation, and peaked near labor. (4) In primary myometrial smooth muscle cell and ex vivo uteri from wild-type mice, lactate decreased interleukin-1ß-induced transcription of key proinflammatory Il1b, Il6, Ccl2, and Pghs2; suppressive effects of lactate were not observed in cells and tissues from GPR81-/- mice. (5) Conversely, proinflammatory gene expression was augmented in the uterus at term in GPR81-/- mice and wild-type mice treated intrauterine with lentiviral-encoded shRNA-GPR81; GPR81 silencing also induced proinflammatory gene transcription in the uterus when labor was induced by endotoxin (lipopolysaccharide). (6) Importantly, administration to pregnant mice of a metabolically stable specific GPR81 agonist, 3,5-dihydroxybenzoic acid, decreased endotoxin-induced uterine inflammation, preterm birth, and associated neonatal mortality. CONCLUSION: Collectively, our data uncover a novel link between the anaerobic glycolysis and the control of uterine inflammation wherein the high levels of lactate produced during labor act on uterine GPR81 to down-regulate key proinflammatory genes. This discovery may represent a novel feedback mechanism to regulate inflammation during labor, and conveys a potential rationale for the use of GPR81 agonists to attenuate inflammation and resulting preterm birth.


Assuntos
Inflamação , Trabalho de Parto/imunologia , Ácido Láctico/imunologia , Miométrio/imunologia , Receptores Acoplados a Proteínas G/genética , Animais , Quimiocina CCL2/efeitos dos fármacos , Quimiocina CCL2/genética , Ciclo-Oxigenase 2/efeitos dos fármacos , Ciclo-Oxigenase 2/genética , Feminino , Hidroxibenzoatos/farmacologia , Imuno-Histoquímica , Técnicas In Vitro , Interleucina-1beta/efeitos dos fármacos , Interleucina-1beta/genética , Interleucina-1beta/farmacologia , Interleucina-6/genética , Trabalho de Parto/metabolismo , Ácido Láctico/metabolismo , Ácido Láctico/farmacologia , Camundongos Knockout , Miométrio/metabolismo , Gravidez , RNA Interferente Pequeno , Receptores Acoplados a Proteínas G/imunologia , Resorcinóis/farmacologia , Útero/imunologia , Útero/metabolismo
4.
Reproduction ; 152(6): R277-R292, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27679863

RESUMO

Inflammation is essential for successful embryo implantation, pregnancy maintenance and delivery. In the last decade, important advances have been made in regard to endogenous, and therefore non-infectious, initiators of inflammation, which can act through the same receptors as pathogens. These molecules are referred to as damage-associated molecular patterns (DAMPs), and their involvement in reproduction has only recently been unraveled. Even though inflammation is necessary for successful reproduction, untimely activation of inflammatory processes can have devastating effect on pregnancy outcomes. Many DAMPs, such as uric acid, high-mobility group box 1 (HMGB1), interleukin (IL)-1 and cell-free fetal DNA, have been associated with pregnancy complications, such as miscarriages, preeclampsia and preterm birth in preclinical models and in humans. However, the specific contribution of alarmins to these conditions is still under debate, as currently there is lack of information on their mechanism of action. In this review, we discuss the role of sterile inflammation in reproduction, including early implantation and pregnancy complications. Particularly, we focus on major alarmins vastly implicated in numerous sterile inflammatory processes, such as uric acid, HMGB1, IL-1α and cell-free DNA (especially that of fetal origin) while giving an overview of the potential role of other candidate alarmins.


Assuntos
Inflamação/fisiopatologia , Complicações na Gravidez/epidemiologia , Feminino , Humanos , Incidência , Gravidez
5.
Biol Reprod ; 95(3): 72, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27512149

RESUMO

Uterine labor requires the conversion of a quiescent (propregnancy) uterus into an activated (prolabor) uterus, with increased sensitivity to endogenous uterotonic molecules. This activation is induced by stressors, particularly inflammation in term and preterm labor. Neuromedin U (NmU) is a neuropeptide known for its uterocontractile effects in rodents. The objective of the study was to assess the expression and function of neuromedin U receptor 2 (NmU-R2) and its ligands NmU and the more potent neuromedin S (NmS) in gestational tissues, and the possible implication of inflammatory stressors in triggering this system. Our data show that NmU and NmS are uterotonic ex vivo in murine tissue, and they dose-dependently trigger labor by acting specifically via NmU-R2. Expression of NmU-R2, NmU, and NmS is detected in murine and human gestational tissues by immunoblot, and the expression of NmS in placenta and of NmU-R2 in uterus increases considerably with gestation age and labor, which is associated with amplified NmU-induced uterocontractile response in mice. NmU- and NmS-induced contraction is associated with increased NmU-R2-coupled Ca++ transients, and Akt and Erk activation in murine primary myometrial smooth muscle cells (mSMCs), which are potentiated with gestational age. NmU-R2 is upregulated in vitro in mSMCs and in vivo in uterus in response to proinflammatory interleukin 1beta (IL1beta), which is associated with increased NmU-induced uterocontractile response and Ca++ transients in murine and human mSMCs; additionally, placental NmS is markedly upregulated in vivo in response to IL1beta. In human placenta at term, immunohistological analysis revealed NmS expression primarily in cytotrophoblasts; furthermore, stimulation with lipopolysaccharide (LPS; Gram-negative endotoxin) markedly upregulates NmS expression in primary human cytotrophoblasts isolated from term placentas. Correspondingly, decidua of women with clinical signs of infection who delivered preterm display significantly higher expression of NmS compared with those without infection. Importantly, in vivo knockdown of NmU-R2 prevents LPS-triggered preterm birth in mice and the associated neonatal mortality. Altogether, our data suggest a critical role for NmU-R2 and its ligands NmU and NmS in preterm labor triggered by infection. We hereby identify NmU-R2 as a relevant target for preterm birth.

6.
Cytokine Growth Factor Rev ; 28: 37-51, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26684042

RESUMO

Preterm birth (PTB) is a leading cause of neonatal mortality and morbidity worldwide, and represents a heavy economic and social burden. Despite its broad etiology, PTB has been firmly linked to inflammatory processes. Pro-inflammatory cytokines are produced in gestational tissues in response to stressors and can prematurely induce uterine activation, which precedes the onset of preterm labor. Of all cytokines implicated, interleukin (IL)-1 has been largely studied, revealing a central role in preterm labor. However, currently approved IL-1-targeting therapies have failed to show expected efficacy in pre-clinical studies of preterm labor. Herein, we (a) summarize animal and human studies in which IL-1 or IL-1-targeting therapeutics are implicated with preterm labor, (b) focus on novel IL-1-targeting therapies and diagnostic tests, and (c) develop the case for commercialization and translation means to hasten their development.


Assuntos
Interleucina-1/imunologia , Trabalho de Parto Prematuro/imunologia , Animais , Feminino , Humanos , Recém-Nascido , Inflamação/imunologia , Terapia de Alvo Molecular , Trabalho de Parto Prematuro/prevenção & controle , Gravidez , Nascimento Prematuro/imunologia , Nascimento Prematuro/prevenção & controle
7.
Microcirculation ; 22(3): 159-67, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25660297

RESUMO

Brain homeostasis is dependent upon the constant perfusion of oxygen and nutrients through CBF. The mechanism responsible to couple CBF to neuronal activity is termed NVC. Ang II, a key player in hypertension, impairs NVC and has been found to be of significant importance in neurodegenerative diseases, such as AD. The presence of Ang II and its metabolites, as well as their receptors have been detected in many areas of the brain. Findings indicate that disruption of NVC by Ang II is mediated by its AT1R and NADPH oxidase (NOX)-dependent ROS production independently of its effect on blood pressure. Although the process causing ROS to interfere with NVC has yet to be elucidated, data label peroxynitrite as the leading ROS to do so. The same common pathway is involved in NVC impairment in AD experimental models. This review investigates recent data concerning the effects of Ang II on NVC with the intent of exploring new treatment perspectives for neurodegenerative diseases such as AD.


Assuntos
Doença de Alzheimer/metabolismo , Doença de Alzheimer/fisiopatologia , Angiotensina II/metabolismo , Acoplamento Neurovascular , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Animais , Modelos Animais de Doenças , Humanos , Hipertensão/metabolismo , Hipertensão/patologia , Hipertensão/fisiopatologia , Hipertensão/terapia , NADPH Oxidases/metabolismo , Receptor Tipo 1 de Angiotensina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA