Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Equity Health ; 22(1): 186, 2023 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-37674167

RESUMEN

Women represent the cornerstone of a family's overall health. Therefore, supporting women's health, particularly in pregnancy, is important to promote public health. Emerging data highlight the contribution of social determinants of health (SDOH) on pregnancy outcomes in understudied, underrepresented, and underreported (U3) populations. Importantly, women are uniquely affected by and more vulnerable to adverse outcomes associated with SDOH. The maternal mortality rate has also increased significantly in the United States, especially among U3 individuals. Factors such as access to safe food, housing and environment, access to education and emergency/health services, and stressors such as interpersonal racism, poverty, unemployment, residential segregation, and domestic violence may make women from U3 populations more vulnerable to adverse reproductive health outcomes. Despite progress in promoting women's health, eliminating social and health disparities in pregnant individuals remains an elusive goal in U3 populations. Moreover, chronic exposure to excessive social/cultural stressors may have a physiologic cost leading to pregnancy complications such as miscarriages, preterm birth, and preeclampsia. Thus, the identification of SDOH-related factors that drive differences in pregnancy-related complications and deaths and the implementation of prevention strategies to address them could reduce disparities in pregnancy-related mortality in U3 populations.


Asunto(s)
Servicios Médicos de Urgencia , Nacimiento Prematuro , Recién Nacido , Embarazo , Femenino , Estados Unidos/epidemiología , Humanos , Determinantes Sociales de la Salud , Salud de la Mujer , Escolaridad
2.
Am J Obstet Gynecol ; 227(4): 571-581, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35662548

RESUMEN

Cannabis is the most commonly used federally illegal drug in the United States and the world, especially among people of reproductive age. In addition, the potency of cannabis products has increased significantly in the past decade. This is concerning because the available evidence suggests an adverse effect of cannabis exposure on male and female reproductive health. Exposure to cannabinoids may have differential impacts on female reproductive health across a woman's lifespan, from preconception to pregnancy, throughout lactation, and during menopause. Moreover, cannabis use has been associated with adverse effects on fetal outcomes and longer-term offspring health and developmental trajectories. Despite the prevalence of cannabis use, there is limited available evidence regarding its safety, especially in regard to reproductive health, pregnancy, and lactation. The biological effects of cannabis are mediated by the endocannabinoid system, and studies have reported the presence of cannabinoid receptors in the male and female reproductive tract, on sperm and the placenta, suggesting that the endocannabinoid system plays a role in regulating reproduction. Cannabis use can affect male and female fertility and has been associated with altered reproductive hormones, menstrual cyclicity, and semen parameters. Use of cannabis in male patients has also been associated with erectile dysfunction, abnormal spermatogenesis, and testicular atrophy. In female patients, cannabis use has been associated with infertility and abnormal embryo implantation and development. The main psychoactive component of cannabis, the delta-9-tetrahydrocannabinol, can also cross the placenta and has been detected in breast milk. Maternal cannabis use during pregnancy and lactation has been associated with adverse effects, including small-for-gestational-age infants, preterm birth, fetal neurodevelopmental consequences, and impaired offspring sociobehavioral and cognitive development. The prevalence of cannabis use for alleviating menopausal symptoms has also increased despite the limited information on its benefits and safety. Given that cannabis use is on the rise, it is critical to understand its impact on reproductive health and offspring developmental outcomes. This is an understudied but timely subject requiring much further information to guide healthcare providers and those interested in conceiving or who are pregnant and lactating, and those at the end of their reproductive time span.


Asunto(s)
Cannabinoides , Cannabis , Drogas Ilícitas , Nacimiento Prematuro , Cannabinoides/efectos adversos , Cannabis/efectos adversos , Dronabinol , Endocannabinoides , Femenino , Hormonas , Humanos , Recién Nacido , Lactancia , Masculino , Embarazo , Receptores de Cannabinoides , Salud Reproductiva , Semillas
3.
J Autoimmun ; 75: 30-38, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27160365

RESUMEN

Placental ischemic disease and adverse pregnancy outcomes are frequently observed in patients with antiphospholipid syndrome (APS). Despite the administration of conventional antithrombotic treatment a significant number of women continue to experience adverse pregnancy outcomes, with uncertain prevention and management. Efforts to develop effective pharmacological strategies for refractory obstetric APS cases will be of significant clinical benefit for both mothers and fetuses. Although the antimalarial drug, hydroxychloroquine (HCQ) is increasingly used to treat pregnant women with APS, little is known about its efficacy and mechanism of action of HCQ. Because complement activation plays a crucial and causative role in placental ischemia and abnormal fetal brain development in APS we hypothesised that HCQ prevents these pregnancy complications through inhibition of complement activation. Using a mouse model of obstetric APS that closely resembles the clinical condition, we found that HCQ prevented fetal death and the placental metabolic changes -measured by proton magnetic resonance spectroscopy in APS-mice. Using 111In labelled antiphospholipid antibodies (aPL) we identified the placenta and the fetal brain as the main organ targets in APS-mice. Using this same method, we found that HCQ does not inhibit aPL binding to tissues as was previously suggested from in vitro studies. While HCQ did not affect aPL binding to fetal brain it prevented fetal brain abnormal cortical development. HCQ prevented complement activation in vivo and in vitro. Complement C5a levels in serum samples from APS patients and APS-mice were lower after treatment with HCQ while the antibodies titres remained unchanged. HCQ prevented not only placental insufficiency but also abnormal fetal brain development in APS. By inhibiting complement activation, HCQ might also be an effective antithrombotic therapy.


Asunto(s)
Síndrome Antifosfolípido/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Activación de Complemento/efectos de los fármacos , Hidroxicloroquina/uso terapéutico , Placenta/efectos de los fármacos , Complicaciones del Embarazo/tratamiento farmacológico , Adulto , Animales , Antimaláricos/uso terapéutico , Síndrome Antifosfolípido/sangre , Encéfalo/anomalías , Encéfalo/embriología , Complemento C3a/análisis , Complemento C5a/análisis , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Placenta/irrigación sanguínea , Embarazo , Complicaciones del Embarazo/sangre , Resultado del Embarazo , Resultado del Tratamiento
4.
Am J Obstet Gynecol ; 215(4): 464.e1-7, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27133010

RESUMEN

BACKGROUND: Congophilia indicates the presence of amyloid protein, which is an aggregate of misfolded proteins, that is implicated in the pathophysiologic condition of preeclampsia. Recently, urinary congophilia has been proposed as a test for the diagnosis and prediction of preeclampsia. OBJECTIVES: The purpose of this study was to determine whether urine congophilia is present in a cohort of women with preeclampsia and in pregnant and nonpregnant women with renal disease. STUDY DESIGN: With the use of a preeclampsia, chronic hypertension, renal disease, and systemic lupus erythematosus cohort, we analyzed urine samples from healthy pregnant control subjects (n = 31) and pregnant women with preeclampsia (n = 23), gestational hypertension (n = 10), chronic hypertension (n = 14), chronic kidney disease; n = 28), chronic kidney disease with superimposed preeclampsia (n = 5), and chronic hypertension and superimposed preeclampsia (n = 12). Samples from nonpregnant control subjects (n = 10) and nonpregnant women with either systemic lupus erythematosus with (n = 25) and without (n = 14) lupus nephritis were analyzed. For each sample, protein concentration was standardized before it was mixed with Congo Red, spotted to nitrocellulose membrane, and rinsed with methanol. The optical density of the residual Congo Red stain was determined; Congo red stain retention was calculated, and groups were compared with the use of the Mann-Whitney test or Kruskal-Wallis analysis of Variance test, as appropriate. RESULTS: Congophilia was increased in urine from women with preeclampsia (median Congo red stain retention, 47%; interquartile range, 22-68%) compared with healthy pregnant control subjects (Congo red stain retention: 16%; interquartile range, 13-21%; P = .002), women with gestational hypertension (Congo red stain retention, 20%; interquartile range, 13-27%; P = .008), or women with chronic hypertension (Congo red stain retention, 17%; interquartile range, 12-28%; P = .01). There were no differences in Congo red retention between pregnant women with chronic hypertension and normal pregnant control subjects (Congo red stain retention, 17% [interquartile range, 12-28%] vs 16% [interquartile range, 13-21%], respectively; P = .72). Congophilia was present in pregnant women with chronic kidney disease (Congo red stain retention, 32%; interquartile range, 14-57%), being similar to values found in women with preeclampsia (P = .22) and for women with chronic kidney disease and superimposed preeclampsia (Congo red stain retention, 57%; [interquartile range, 29-71%; P = .18). Nonpregnant women with lupus nephritis had higher congophilia levels compared with nonpregnant female control subjects (Congo red stain retention, 38% [interquartile range, 17-73%] vs 9% [7-11%], respectively; P < .001) and nonpregnant women with systemic lupus erythematosus without nephritis (Congo red stain retention, 38% [interquartile range, 17-73%] vs 13% [interquartile range, 11-17%], respectively; P = .001). A significant positive correlation was observed between congophilia and protein:creatinine ratio (Spearman rank correlations, 0.702; 95% confidence interval, 0.618-0.770; P < .001). CONCLUSION: This study confirms that women with preeclampsia and chronic kidney disease without preeclampsia have elevated urine congophilia levels compared with healthy pregnant women. Nonpregnant women with lupus nephritis also have elevated urine congophilia levels compared with healthy control subjects. An elevated Congo Red stain retention may not be able to differentiate between these conditions; further research is required to explore the use of congophilia in clinical practice.


Asunto(s)
Proteínas Amiloidogénicas/orina , Nefritis Lúpica/orina , Preeclampsia/orina , Insuficiencia Renal Crónica/orina , Adulto , Estudios de Casos y Controles , Enfermedad Crónica , Colorantes , Rojo Congo , Femenino , Humanos , Hipertensión/orina , Embarazo , Proteinuria/orina , Adulto Joven
5.
Biochim Biophys Acta ; 1842(1): 107-15, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24184716

RESUMEN

Premature babies are particularly vulnerable to brain injury. In this study we focus on cortical brain damage associated with long-term cognitive, behavioral, attentional or socialization deficits in children born preterm. Using a mouse model of preterm birth (PTB), we demonstrated that complement component C5a contributes to fetal cortical brain injury. Disruption of cortical dendritic and axonal cytoarchitecture was observed in PTB-mice. Fetuses deficient in C5aR (-/-) did not show cortical brain damage. Treatment with antibody anti-C5, that prevents generation of C5a, also prevented cortical fetal brain injury in PTB-mice. C5a also showed a detrimental effect on fetal cortical neuron development and survival in vitro. Increased glutamate release was observed in cortical neurons in culture exposed to C5a. Blockade of C5aR prevented glutamate increase and restored neurons dendritic and axonal growth and survival. Similarly, increased glutamate levels - measured by (1)HMRS - were observed in vivo in PTB-fetuses compared to age-matched controls. The blockade of glutamate receptors prevented C5a-induced abnormal growth and increased cell death in isolated fetal cortical neurons. Simvastatin and pravastatin prevented cortical fetal brain developmental and metabolic abnormalities -in vivo and in vitro. Neuroprotective effects of statins were mediated by Akt/PKB signaling pathways. This study shows that complement activation plays a crucial role in cortical fetal brain injury in PTL and suggests that complement inhibitors and statins might be good therapeutic options to improve neonatal outcomes in preterm birth.


Asunto(s)
Corteza Cerebral/metabolismo , Activación de Complemento , Complemento C5a/metabolismo , Neuronas/metabolismo , Nacimiento Prematuro/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Células Cultivadas , Corteza Cerebral/anomalías , Corteza Cerebral/efectos de los fármacos , Complemento C5a/antagonistas & inhibidores , Complemento C5a/genética , Femenino , Feto , Regulación de la Expresión Génica , Ácido Glutámico/metabolismo , Humanos , Recién Nacido , Ratones , Modelos Animales , Neuronas/efectos de los fármacos , Neuronas/patología , Pravastatina/farmacología , Embarazo , Nacimiento Prematuro/genética , Nacimiento Prematuro/patología , Receptor de Anafilatoxina C5a/deficiencia , Receptor de Anafilatoxina C5a/genética , Receptores de Glutamato/genética , Receptores de Glutamato/metabolismo , Transducción de Señal , Simvastatina/farmacología
6.
Mol Hum Reprod ; 20(6): 579-89, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24623738

RESUMEN

Preterm birth (PTB) is a major public health problem, with a global prevalence of 9.6% and over a million annual neonatal deaths. In a mouse model of preterm labor (PTL) induced by intravaginal administration of a subclinical dose of lipopolysaccharide (LPS), we previously demonstrated that LPS ascends to the cervix, inducing complement activation, cervical remodeling and PTL. Here we show that complement activation also plays a role in myometrial contractions during PTL in this model. Increased levels of C5a were detected in the myometrium of LPS-treated mice but not in age-matched control or term myometrium. Human and mouse myometrium incubated with C5a showed increased frequency of contractions and expression of connexin 43, suggesting that C5a is an uterotonic molecule. Statins, which showed beneficial effects in preventing complement-mediated pregnancy complications, prevented cervical remodeling, myometrial contractions and PTL in the LPS model. The protective effects of statins in PTL were associated with increased synthesis, expression and activity of heme oxygenase (HO-1) in myometrium and cervix. Coadministration of HO-1 inhibitor tin-protoporphyrin-IX with pravastatin abrogated the protective effects of pravastatin on cervical remodeling and myometrial contractions leading to PTB. In addition, pravastatin inhibited complement activation in the cervix by increasing the synthesis and expression of complement inhibitor decay-accelerating factor. This study in mice suggests that statins might be useful to prevent PTL in humans. Clinical trials in humans are needed and if these results are confirmed, they may form the basis for a new clinical approach to prevent PTB.


Asunto(s)
Cuello del Útero/efectos de los fármacos , Complemento C5a/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Hemo-Oxigenasa 1/genética , Miometrio/efectos de los fármacos , Trabajo de Parto Prematuro/prevención & control , Pravastatina/farmacología , Adulto , Animales , Antígenos CD55/genética , Antígenos CD55/metabolismo , Cuello del Útero/metabolismo , Cuello del Útero/fisiopatología , Complemento C5a/farmacología , Conexina 43/genética , Conexina 43/metabolismo , Femenino , Regulación de la Expresión Génica , Hemo-Oxigenasa 1/metabolismo , Humanos , Lipopolisacáridos , Masculino , Metaloporfirinas/farmacología , Ratones , Ratones Endogámicos C57BL , Miometrio/metabolismo , Miometrio/fisiopatología , Trabajo de Parto Prematuro/inducido químicamente , Trabajo de Parto Prematuro/genética , Trabajo de Parto Prematuro/metabolismo , Embarazo , Protoporfirinas/farmacología , Contracción Uterina/efectos de los fármacos
7.
Birth Defects Res ; 116(2): e2313, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38348550

RESUMEN

BACKGROUND: The effects of climate and environmental changes (CEC) are being felt globally and will worsen over the next decade unless significant changes are made on a global level. Climate change is having serious consequences for health, particularly for vulnerable women and their offspring and less resilient individuals in communities with socioeconomic inequalities. To protect human health from CEC effects, efforts need to be directed toward building resilience strategies. Building political and economic power, as well as directly addressing CEC-related challenges, are critical components of climate resilience. Effective communication and tailored methods to engage women in preventive strategies are also necessary to ameliorate the deleterious effects of CEC on women's health. Furthermore, women from marginalized communities face more CEC-associated challenges. CONCLUSIONS: Therefore, effective policies and programs targeting these at-risk populations-are crucial to improve the overall state of global health. In closing, it is time to increase awareness of the effects of CECs on women's health and their transgenerational effects in order to ensure that all people, regardless of race, ethnicity, education and income are protected from the detrimental effects of CECs.


Asunto(s)
Clorambucilo , Salud de la Mujer , Lactante , Embarazo , Humanos , Femenino , Etopósido , Lomustina , Inequidades en Salud
8.
J Exp Med ; 204(5): 1049-56, 2007 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-17438064

RESUMEN

We describe a mouse model of fetal loss in factor V Leiden (FvL) mothers in which fetal loss is triggered when the maternal prothrombotic state coincides with fetal gene defects that reduce activation of the protein C anticoagulant pathway within the placenta. Fetal loss is caused by disruption of placental morphogenesis at the stage of labyrinth layer formation and occurs in the absence of overt placental thrombosis, infarction, or perfusion defects. Platelet depletion or elimination of protease-activated receptor 4 (Par4) from the mother allows normal placentation and prevents fetal loss. These findings establish a cause-effect relationship for the observed epidemiologic association between maternal FvL status and fetal loss and identify fetal gene defects as risk modifiers of pregnancy failure in prothrombotic mothers. Pregnancy failure is mediated by Par4-dependent activation of maternal platelets at the fetomaternal interface and likely involves a pathogenic pathway independent of occlusive thrombosis. Our results further demonstrate that the interaction of two given thrombosis risk factors produces markedly disparate consequences on disease manifestation (i.e., thrombosis or pregnancy loss), depending on the vascular bed in which this interaction occurs.


Asunto(s)
Resistencia a la Proteína C Activada/complicaciones , Plaquetas/metabolismo , Modelos Animales de Enfermedad , Factor V/genética , Muerte Fetal/etiología , Enfermedades Fetales/genética , Placenta/patología , Resistencia a la Proteína C Activada/genética , Animales , Femenino , Muerte Fetal/patología , Ratones , Ratones Endogámicos C57BL , Placenta/irrigación sanguínea , Mutación Puntual/genética , Embarazo , Resultado del Embarazo/genética , Receptores de Trombina/metabolismo , Factores de Riesgo , Trombomodulina/genética
9.
J Exp Med ; 203(9): 2165-75, 2006 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-16923853

RESUMEN

Immune mechanisms have been implicated in placental dysfunction in patients with recurrent miscarriages and intrauterine growth restriction (IUGR), but the mediators are undefined. Here we show that complement activation, particularly C5a, is a required intermediary event in the pathogenesis of placental and fetal injury in an antibody-independent mouse model of spontaneous miscarriage and IUGR, and that complement activation causes dysregulation of the angiogenic factors required for normal placental development. Pregnancies complicated by miscarriage or growth restriction were characterized by inflammatory infiltrates in placentas, functional deficiency of free vascular endothelial growth factor (VEGF), elevated levels of soluble VEGF receptor 1 (sVEGFR-1, also known as sFlt-1; a potent anti-angiogenic molecule), and defective placental development. Inhibition of complement activation in vivo blocked the increase in sVEGFR-1 and rescued pregnancies. In vitro stimulation of monocytes with products of the complement cascade directly triggered release of sVEGFR-1, which sequesters VEGF. These studies provide the first evidence linking the complement system to angiogenic factor imbalance associated with placental dysfunction, and identify a new effector of immune-triggered pregnancy complications.


Asunto(s)
Aborto Espontáneo/inmunología , Inductores de la Angiogénesis/metabolismo , Activación de Complemento/fisiología , Retardo del Crecimiento Fetal/inmunología , Insuficiencia Placentaria/inmunología , Animales , Complemento C5a/inmunología , Embrión de Mamíferos/inmunología , Embrión de Mamíferos/patología , Embrión de Mamíferos/fisiología , Femenino , Feto/inmunología , Feto/patología , Humanos , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos , Monocitos/metabolismo , Placenta/citología , Placenta/inmunología , Placenta/patología , Placenta/fisiología , Enfermedades Placentarias/inmunología , Enfermedades Placentarias/patología , Embarazo , Complicaciones del Embarazo/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/inmunología
10.
Am J Pathol ; 179(2): 838-49, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21801872

RESUMEN

Inflammation is frequently linked to preterm delivery (PTD). Here, we tested the hypothesis that complement activation plays a role in cervical remodeling and PTD. We studied two mouse models of inflammation-induced PTD. The first model was induced by vaginal administration of lipopolysaccharide (LPS) and the second one by administration of progesterone antagonist RU486. Increased cervical C3 deposition and macrophages infiltration and increased serum C3adesArg and C5adesArg levels were observed in both models when compared to gestational age matched controls. A significant increase in collagen degradation, matrix metalloproteinase 9 (MMP-9) activity and tissue distensibility was observed in the cervix in both models. Mice deficient in complement receptor C5a did not show increased MMP-9 activity and cervical remodeling and did not deliver preterm in response to LPS or RU486, suggesting a role for C5aR in the cervical changes that precede PTD. In vitro studies show that macrophages release MMP-9 in response to C5a. Progesterone diminished the amount of C5aR on the macrophages surface, inhibited the release of MMP-9 and prevented PTD. In addition, macrophages depletion also prevented cervical remodeling and PTD in LPS-treated mice. Our studies show that C5a-C5aR interaction is required for MMP-9 release from macrophages, and the cervical remodeling that leads to PTD. Complement inhibition and supplementation with progesterone may be good therapeutic options to prevent this serious pregnancy complication.


Asunto(s)
Cuello del Útero/patología , Metaloproteasas/metabolismo , Animales , Activación de Complemento , Complemento C3/metabolismo , Complemento C5/metabolismo , Femenino , Inflamación , Lipopolisacáridos/metabolismo , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Embarazo , Preñez , Nacimiento Prematuro/metabolismo , Nacimiento Prematuro/prevención & control
11.
J Pathol ; 225(4): 502-11, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21688269

RESUMEN

Recurrent miscarriage, fetal growth restriction and intrauterine fetal death are frequently occurring complications of pregnancy in patients with systemic lupus erythaematosus (SLE) and antiphospholipid syndrome (APS). Murine models show that complement activation plays a pivotal role in antiphospholipid antibody-mediated pregnancy morbidity, but the exact pathways of complement activation and their potential role in human pregnancy are insufficiently understood. We hypothesized that the classical pathway would play a major role in inducing fetal loss. Pregnant C57BL/6 mice and mice deficient in C1q and factor D were injected with antiphospholipid antibodies or normal human IgG. Mouse placentas were subsequently stained with an anti-C4 antibody and anti-normal human IgG to determine the presence of classical complement activation and IgG binding. Findings in mice were validated in 88 human placentae from 83 women (SLE and APS cases versus controls), which were immunohistochemically stained for C4d, C1q, properdin and MBL. Staining patterns were compared to pregnancy outcome. In murine placentae of mice pretreated with antiphospholipid antibodies, increased C4 deposition was observed, which was associated with adverse fetal outcome but not with IgG binding. In humans, diffuse C4d staining at the feto-maternal interface was present almost exclusively in patients with SLE and/or APS (p < 0.001) and was related to intrauterine fetal death (p = 0.03). Our data show that presence of C4d in murine and human placentae is strongly related to adverse fetal outcome in the setting of SLE and APS. The excessive deposition of C4d supports the concept of severe autoantibody-mediated injury at the fetal-maternal interface. We suggest C4d as a potential biomarker of autoantibody-mediated fetal loss in SLE and APS.


Asunto(s)
Anticuerpos Antifosfolípidos/inmunología , Síndrome Antifosfolípido/inmunología , Activación de Complemento/inmunología , Complemento C1q/inmunología , Muerte Fetal/inmunología , Adulto , Animales , Anticuerpos Antifosfolípidos/administración & dosificación , Síndrome Antifosfolípido/complicaciones , Síndrome Antifosfolípido/patología , Biomarcadores , Complemento C1q/deficiencia , Complemento C4/metabolismo , Modelos Animales de Enfermedad , Femenino , Muerte Fetal/inducido químicamente , Edad Gestacional , Humanos , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Placenta/inmunología , Placenta/metabolismo , Placenta/patología , Embarazo , Resultado del Embarazo
12.
J Immunol ; 185(7): 4420-9, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20810993

RESUMEN

Fetal trophoblast cells invading the decidua in the early phase of pregnancy establish complex interaction with the maternal extracellular matrix. We discovered that C1q was widely distributed in human decidual stroma in the absence of C4 and C3 and was actively synthesized by migrating extravillous trophoblasts. The cells expressed the messages for the three chains of C1q and secreted this complement component that interacted with the proteins of the decidual extracellular matrix. Solid phase-bound C1q promoted trophoblast adhesion and migration, and cell binding to C1q resulted in activation of ERK1/2 MAPKs. Ab inhibition experiments showed that the receptors for the globular head of C1q/p33 and α(4)ß(1) integrin were both involved in this process and were colocalized on the cell surface following binding of C1q to trophoblasts. We also found that C1q(-/-) mice manifested increased frequency of fetal resorption, reduced fetal weight, and smaller litter sizes compared with wild-type mice. C1q deficiency was associated with impaired labyrinth development and decidual vessel remodeling. Collectively, these data suggest that C1q plays an important role in promoting trophoblast invasion of decidua and that defective local production of C1q may be involved in pregnancy disorders, such as pre-eclampsia, characterized by poor trophoblast invasion.


Asunto(s)
Quimiotaxis de Leucocito/fisiología , Complemento C1q/metabolismo , Placentación/inmunología , Trofoblastos/metabolismo , Animales , Adhesión Celular/inmunología , Complemento C1q/inmunología , Femenino , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Preeclampsia/inmunología , Embarazo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Trofoblastos/inmunología
13.
Obstet Gynecol ; 139(1): 3-8, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34727554

RESUMEN

Three coronavirus disease 2019 (COVID-19) vaccines have been authorized for use in the United States; specifically, the Pfizer-BioNTech, Moderna, and Johnson & Johnson-Janssen COVID-19 vaccines were granted emergency use authorization by the U.S. Food and Drug Administration in late 2020 and early 2021. Vaccination coverage and intent among adults are lowest among those aged 18-39 years and among females in particular. In females of reproductive age, enthusiasm for receiving a COVID-19 vaccine may be negatively affected by claims currently circulating widely on diverse social media platforms regarding the vaccines adversely affecting fertility and pregnancy. Yet it is important to note that these claims are anecdotal in nature and not supported by the available scientific evidence. It is also imperative that the effects of COVID-19 vaccine on reproductive health are clarified. Herein, we discuss the existing scientific data supporting COVID-19 vaccine safety and efficacy in people who are planning to conceive or who are pregnant or lactating and highlight the importance of COVID-19 vaccination in females of reproductive age.


Asunto(s)
Vacunas contra la COVID-19/administración & dosificación , COVID-19/prevención & control , Atención Preconceptiva , Complicaciones Infecciosas del Embarazo/prevención & control , Atención Prenatal , Infección Puerperal/prevención & control , SARS-CoV-2 , Eficacia de las Vacunas , Adolescente , Adulto , Femenino , Humanos , Lactancia , Embarazo , Adulto Joven
14.
Am J Reprod Immunol ; 87(1): e13508, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34716735

RESUMEN

Recurrent pregnancy loss (RPL) is one of the most complex and challenging scenarios in reproductive medicine. New theories about the mechanisms behind RPL have recently emerged, highlighting the multifactorial nature of this serious pregnancy complication.  Unfortunately, these preclinical observations are rarely validated in the human scenario, where treatment remains ineffective and empirical. New technologies such as organoids, organ-on-a-chip, and 3D printing can be used to characterize the molecular cross talk between the uterine environment with its unique inflammatory cells and the developing embryo. Understanding the mechanisms behind RPL and identifying mediators and effectors and validating these targets for prevention and therapy in humans will have a profound impact on women's health.


Asunto(s)
Aborto Habitual/etiología , Útero/patología , Aborto Habitual/patología , Animales , Femenino , Humanos , Embarazo , Factores de Riesgo
15.
Metab Syndr Relat Disord ; 20(5): 251-254, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35384734

RESUMEN

The prevalence of obesity is ∼40% in the United States, and the prepregnancy prevalence of obesity in females is ∼30%. This has in part fueled an increase in metabolic syndrome (MetS) among females who are currently pregnant, have been pregnant, or are planning to become pregnant. Importantly, MetS in pregnancy is associated with increased pregnancy complications. Moreover, MetS in pregnancy may have long-lasting adverse cardiovascular and metabolic health implications for the mother and her offspring. To complicate matters, many adverse pregnancy outcomes seem to increase the risk of MetS in the mother after pregnancy. Herein, we describe the potential mechanisms behind the intersection of MetS, adverse pregnancy outcomes, and subsequent long-term disease in the mother and offspring. Because MetS is a cluster of coexisting conditions, it is challenging to identify mediators that can serve as biomarkers for early diagnosis and targets for MetS prevention and therapy.


Asunto(s)
Diabetes Gestacional , Síndrome Metabólico , Complicaciones del Embarazo , Femenino , Humanos , Síndrome Metabólico/complicaciones , Síndrome Metabólico/diagnóstico , Síndrome Metabólico/epidemiología , Madres , Obesidad/complicaciones , Obesidad/epidemiología , Obesidad/terapia , Embarazo , Complicaciones del Embarazo/diagnóstico , Complicaciones del Embarazo/epidemiología , Resultado del Embarazo/epidemiología , Factores de Riesgo
16.
J Clin Invest ; 118(10): 3453-61, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18802482

RESUMEN

Women with antiphospholipid syndrome (APS), a condition characterized by the presence of antiphospholipid antibodies (aPL), often suffer pregnancy-related complications, including miscarriage. We have previously shown that C5a induction of tissue factor (TF) expression in neutrophils contributes to respiratory burst, trophoblast injury, and pregnancy loss in mice treated with aPL. Here we analyzed how TF contributes to neutrophil activation and trophoblast injury in this model. Neutrophils from aPL-treated mice expressed protease-activated receptor 2 (PAR2), and stimulation of this receptor led to neutrophil activation, trophoblast injury, and fetal death. An antibody specific for human TF that has little impact on coagulation, but potently inhibits TF/Factor VIIa (FVIIa) signaling through PAR2, inhibited aPL-induced neutrophil activation in mice that expressed human TF. Genetic deletion of the TF cytoplasmic domain, which allows interaction between TF and PAR2, reduced aPL-induced neutrophil activation in aPL-treated mice. Par2-/- mice treated with aPL exhibited reduced neutrophil activation and normal pregnancies, which indicates that PAR2 plays an important role in the pathogenesis of aPL-induced fetal injury. We also demonstrated that simvastatin and pravastatin decreased TF and PAR2 expression on neutrophils and prevented pregnancy loss. Our results suggest that TF/FVIIa/PAR2 signaling mediates neutrophil activation and fetal death in APS and that statins may be a good treatment for women with aPL-induced pregnancy complications.


Asunto(s)
Síndrome Antifosfolípido/complicaciones , Síndrome Antifosfolípido/fisiopatología , Modelos Animales de Enfermedad , Factor VIIa/metabolismo , Muerte Fetal/etiología , Activación Neutrófila , Receptor PAR-2/metabolismo , Tromboplastina/metabolismo , Animales , Anticuerpos Antifosfolípidos/inmunología , Anticuerpos Antifosfolípidos/farmacología , Síndrome Antifosfolípido/inmunología , Femenino , Muerte Fetal/inmunología , Muerte Fetal/prevención & control , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inmunoglobulina G/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Fagocitosis/efectos de los fármacos , Embarazo , Especies Reactivas de Oxígeno/metabolismo , Receptor PAR-1/deficiencia , Receptor PAR-2/genética , Estallido Respiratorio/efectos de los fármacos , Transducción de Señal , Simvastatina/farmacología , Tromboplastina/genética , Tromboplastina/inmunología
17.
Blood ; 113(17): 4101-9, 2009 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-19234141

RESUMEN

Pregnancy loss and intrauterine growth restriction (IUGR) are serious pregnancy complications, and the triggers and mediators of placental and fetal damage are not completely understood. Using a mouse model of recurrent spontaneous miscarriages (DBA/2-mated CBA/J mice) that shares features with human recurrent miscarriage and fetal growth restriction, we identified tissue factor (TF) as an essential participating factor in placental and fetal injury. We have previously shown that C5a releases antiangiogenic molecule sFlt-1 in monocytes that causes defective placental development and fetal death in DBA/2-mated CBA/J mice. In this study, we found that TF not only activates the coagulation pathway, but it also mediates sFlt-1 release in monocytes causing defective placental development and fetal death. Blockade of TF with a monoclonal antibody inhibited sFlt-1 release, prevented the pathological activation of the coagulation pathway, restored placental blood flow, prevented placental oxidative stress, and rescued pregnancies. We also demonstrated that pravastatin, by down-regulating TF expression on monocytes and trophoblasts, prevented placental damage and protected pregnancies in DBA/2-mated CBA/J mice. These studies indicate that TF is an important mediator in fetal death and growth restriction and that statins may be a good treatment for women with recurrent miscarriages and IUGR.


Asunto(s)
Aborto Espontáneo/metabolismo , Aborto Espontáneo/prevención & control , Placenta/metabolismo , Pravastatina/farmacología , Tromboplastina/metabolismo , Animales , Animales Recién Nacidos/lesiones , Animales Recién Nacidos/metabolismo , Anticoagulantes/farmacología , Antitrombina III/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Recién Nacido , Masculino , Ratones , Monocitos/efectos de los fármacos , Monocitos/metabolismo , Óxidos de Nitrógeno/metabolismo , Estrés Oxidativo , Placenta/irrigación sanguínea , Embarazo , Unión Proteica , Trombina/metabolismo , Trofoblastos/efectos de los fármacos , Trofoblastos/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
18.
Blood ; 114(8): 1675-83, 2009 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-19535796

RESUMEN

Using different mouse monoclonal and human antiphospholipid (aPL) antibodies, we developed a new animal model of renal injury that shares many features with thrombotic microangiopathy (TMA). We found that more than 1 mechanism/signaling pathway is involved in glomerular injury induced by aPL antibodies in this model. Both complement-dependent and complement-independent pathways were identified that lead to glomerular endothelial cell damage and renal function impairment. We also found that C5a-C5aR interaction is a crucial step for the activation of the coagulation cascade and glomerular injury induced by complement-activating antibodies. In addition, our studies demonstrated complement-independent mechanisms in which reactivity with beta(2) glycoprotein I (beta2GPI) plays an important role in aPL-induced glomerular damage and renal failure. Independently of the mechanism responsible for aPL-induced TMA, mice that express low levels of tissue factor (TF) were protected from glomerular injury. That genetic reduction of TF prevents renal injury induced by different aPL antibodies indicates that TF is a common mediator of glomerular damage and a possible target for selective pharmacologic intervention. Treatment with pravastatin, which down-regulates glomerular TF synthesis, prevents aPL-induced TMA in this mouse model, thus emphasizing that targeting TF might be a good therapeutic intervention in patients with TMA.


Asunto(s)
Anticuerpos Antifosfolípidos/efectos adversos , Tromboplastina/fisiología , Trombosis/etiología , Adulto , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Reacciones Antígeno-Anticuerpo/inmunología , Modelos Animales de Enfermedad , Femenino , Humanos , Glomérulos Renales/inmunología , Glomérulos Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Microvasos/patología , Persona de Mediana Edad , Insuficiencia Renal/etiología , Insuficiencia Renal/genética , Insuficiencia Renal/inmunología , Insuficiencia Renal/metabolismo , Tromboplastina/genética , Tromboplastina/metabolismo , Trombosis/genética , Trombosis/metabolismo , Trombosis/patología
19.
Nat Med ; 10(11): 1222-6, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15489858

RESUMEN

The antiphospholipid syndrome (APS) is defined by thrombosis and recurrent pregnancy loss in the presence of antiphospholipid (aPL) antibodies and is generally treated with anticoagulation therapy. Because complement activation is essential and causative in aPL antibody-induced fetal injury, we hypothesized that heparin protects pregnant APS patients from complications through inhibition of complement. Treatment with heparin (unfractionated or low molecular weight) prevented complement activation in vivo and in vitro and protected mice from pregnancy complications induced by aPL antibodies. Neither fondaparinux nor hirudin, other anticoagulants, inhibited the generation of complement split products or prevented pregnancy loss, demonstrating that anticoagulation therapy is insufficient protection against APS-associated miscarriage. Our data indicate that heparins prevent obstetrical complications in women with APS because they block activation of complement induced by aPL antibodies targeted to decidual tissues, rather than by their anticoagulant effects.


Asunto(s)
Anticuerpos Antifosfolípidos/inmunología , Síndrome Antifosfolípido/inmunología , Activación de Complemento/efectos de los fármacos , Muerte Fetal/inmunología , Heparina/farmacología , Animales , Síndrome Antifosfolípido/tratamiento farmacológico , Western Blotting , Cromatografía de Afinidad , Activación de Complemento/inmunología , Electroforesis en Gel de Poliacrilamida , Femenino , Heparina/uso terapéutico , Inmunohistoquímica , Ratones , Ratones Endogámicos BALB C , Embarazo
20.
Vascul Pharmacol ; 137: 106824, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33249273

RESUMEN

BACKGROUND: Uteroplacental vascular dysfunction, characterized by diminished uterine artery (UtA) blood flow in the second trimester is a clinically useful predictor of the further development of preeclampsia, fetal growth restriction and stillbirth. Efforts to develop effective treatments to protect pregnancies with abnormal UtA Dopplers would be of significant clinical benefit for mothers and their fetuses. OBJECTIVE: The aim of this pilot non randomized control study was to use pravastatin +L-arginine to improve uteroplacental haemodynamics and prevent adverse maternal and neonatal outcomes in women with abnormal Dopplers and high risk for developing adverse pregnancy outcomes. STUDY DESIGN: This study was performed between 2015 and 2018. All women received primary care at OB/GYN Polyclinic Jurisic and Narodni Front University Hospital, University of Belgrade Medical School, Serbia. Approval for investigational drug use was obtained and all women gave informed consent. 10 pregnant women with a poor obstetric history that developed uteroplacental dysfunction (UtA pulsatility index (PI) above the 95th percentile and notching) at 20.5 weeks IQR [17.7-22] gave consent to be treated daily with pravastatin (40 mg) and L-arginine (1.5 g) to improve placental blood flow and pregnancy outcomes. 5 women remained untreated after diagnosis at 21 weeks [20-22] (control group). Due to presence of risk factors for pregnancy complications, close maternal and fetal monitoring was undertaken in all patients. Doppler examinations were performed to monitor changes in placental vascular resistance and fetal well-being and growth. RESULTS: PRAV+L-arginine improved uteroplacental haemodynamics, increased fetal growth and prevented early onset preeclampsia leading to delivery close to term (delivery date: median 38 weeks, IQR[36.5-39]) and appropriate weight for gestational age compared to controls, in which placental blood flow did not improve and 2 women developed severe early onset preeclampsia. Neonates from the control group were born preterm (25 weeks IQR[23.5-25]), growth restricted and spent several months at NICU. Two neonates died due to prematurity-associated complications. PRAV+L-arginine treatment prolonged pregnancies for 4.1 months, compared to 26 days in the untreated group, preventing neonatal complications associated with prematurity. The infants are now 1-3 years old and show normal growth and development. CONCLUSION: This study describes the successful management with pravastatin+L-arginine of 10 pregnant patients with uteroplacental vascular dysfunction and high risk of adverse maternal and fetal outcomes. A larger study is being organized to confirm these observations.


Asunto(s)
Arginina/uso terapéutico , Retardo del Crecimiento Fetal/tratamiento farmacológico , Hemodinámica/efectos de los fármacos , Circulación Placentaria/efectos de los fármacos , Insuficiencia Placentaria/tratamiento farmacológico , Pravastatina/uso terapéutico , Preeclampsia/prevención & control , Adulto , Arginina/efectos adversos , Quimioterapia Combinada , Femenino , Retardo del Crecimiento Fetal/diagnóstico por imagen , Retardo del Crecimiento Fetal/fisiopatología , Humanos , Nacimiento Vivo , Proyectos Piloto , Insuficiencia Placentaria/diagnóstico por imagen , Insuficiencia Placentaria/fisiopatología , Pravastatina/efectos adversos , Preeclampsia/diagnóstico , Preeclampsia/fisiopatología , Embarazo , Factores de Tiempo , Resultado del Tratamiento , Ultrasonografía Doppler , Ultrasonografía Prenatal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA