Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-34164564

RESUMO

The coronavirus SARS-CoV-2 is cause of a global pandemic of a pneumonia-like disease termed Coronavirus Disease 2019 (COVID-19). COVID-19 presents a high mortality rate, estimated at 3.4%. More than 1 out of 4 hospitalized COVID-19 patients require admission to an Intensive Care Unit (ICU) for respiratory support, and a large proportion of these ICU-COVID-19 patients, between 17% and 46%, have died. In these patients COVID-19 infection causes an inflammatory response in the lungs that can progress to inflammation with cytokine storm, Acute Lung Injury (ALI), Acute Respiratory Distress Syndrome (ARDS), thromboembolic events, disseminated intravascular coagulation, organ failure, and death. Mesenchymal Stem Cells (MSCs) are potent immunomodulatory cells that recognize sites of injury, limit effector T cell reactions, and positively modulate regulatory cell populations. MSCs also stimulate local tissue regeneration via paracrine effects inducing angiogenic, anti-fibrotic and remodeling responses. MSCs can be derived in large number from the Umbilical Cord (UC). UC-MSCs, utilized in the allogeneic setting, have demonstrated safety and efficacy in clinical trials for a number of disease conditions including inflammatory and immune-based diseases. UC-MSCs have been shown to inhibit inflammation and fibrosis in the lungs and have been utilized to treat patients with severe COVID-19 in pilot, uncontrolled clinical trials, that reported promising results. UC-MSCs processed at our facility have been authorized by the FDA for clinical trials in patients with an Alzheimer's Disease, and in patients with Type 1 Diabetes (T1D). We hypothesize that UC-MSC will also exert beneficial therapeutic effects in COVID-19 patients with cytokine storm and ARDS. We propose an early phase controlled, randomized clinical trial in COVID-19 patients with ALI/ARDS. Subjects in the treatment group will be treated with two doses of UC-MSC (l00 × 106 cells). The first dose will be infused within 24 hours following study enrollment. A second dose will be administered 72 ± 6 hours after the first infusion. Subject in the control group will receive infusion of vehicle (DPBS supplemented with 1% HSA and 70 U/kg unfractionated Heparin, delivered IV) following the same timeline. Subjects will be evaluated daily during the first 6 days, then at 14, 28, 60, and 90 days following enrollment (see Schedule of Assessment for time window details). Safety will be determined by adverse events (AEs) and serious adverse events (SAEs) during the follow-up period. Efficacy will be defined by clinical outcomes, as well as a variety of pulmonary, biochemical and immunological tests. Success of the current study will provide a framework for larger controlled, randomized clinical trials and a means of accelerating a possible solution for this urgent but unmet medical need. The proposed early phase clinical trial will be performed at the University of Miami (UM), in the facilities of the Diabetes Research Institute (DRI), UHealth Intensive Care Unit (ICU) and the Clinical Translational Research Site (CTRS) at the University of Miami Miller School of Medicine and at the Jackson Memorial Hospital (JMH).

2.
J Reprod Immunol ; 114: 58-64, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26257082

RESUMO

Preeclampsia is a unique pregnancy disorder whose patho-physiology is initiated early in gestation, while clinical manifestations typically occur in mid-to-late pregnancy. Thus, prevention should optimally be initiated in early gestation. The intimate interaction between PIF, secreted early by viable embryos, and its host-mother provides insight into putative mechanisms of preeclampsia prevention. PIF is instrumental at the two critical events underlying preeclampsia. At first, shallow implantation leads to impaired placentation, oxidative stress, protein misfolding, and endothelial dysfunction. Later in gestation, hyper-oxygenation due to overflow of maternally derived oxygenated blood compromises the placenta. The first is likely involved in early preeclampsia occurrence due to reduced effectiveness of trophoblast/uterus interaction. The latter is observed with later-onset preeclampsia, caused by a breakdown in placental blood flow regulation. We reported that 1. PIF promotes implantation, endometrium receptivity, trophoblast invasion and increases pro-tolerance trophoblastic HLA-G expression and, 2. PIF protects against oxidative stress and protein misfolding, interacting with specific targets in embryo, 3. PIF regulates systemic immunity to reduce oxidative stress. Using PIF as an early preventative preeclampsia intervention could ameliorate or even prevent the disease, whose current main solution is early delivery.


Assuntos
Implantação do Embrião/imunologia , Estresse Oxidativo/imunologia , Pré-Eclâmpsia/imunologia , Proteínas da Gravidez/imunologia , Trofoblastos/imunologia , Feminino , Humanos , Pré-Eclâmpsia/prevenção & controle , Gravidez
3.
Pharmacol Ther ; 156: 10-25, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26546485

RESUMO

Embryonic-maternal interaction from the earliest stages of gestation has a key, sustained role in neurologic development, persisting into adulthood. Early adverse events may be detrimental in adulthood. Protective factors present during gestation could significantly impact post-natal therapy. The role of PreImplantation Factor (PIF) within this context is herein examined. Secreted by viable early embryos, PIF establishes effective embryonic-maternal communication and exerts essential trophic and protective roles by reducing oxidative stress and protein misfolding and by blunting the nocive let-7 microRNA related pathway. PIF's effects on systemic immunity lead to comprehensive immune modulation, not immune suppression. We examine PIF's role in protecting embryos from adverse maternal environment, which can lead to neurological disorders that may only manifest post-nataly: Synthetic PIF successfully translates endogenous PIF features in both pregnant and non-pregnant clinically relevant models. Specifically PIF has neuroprotective effects in neonatal prematurity. In adult relapsing-remitting neuroinflammation, PIF reverses advanced paralysis while promoting neurogenesis. PIF reversed Mycobacterium smegmatis induced brain infection. In graft-vs.-host disease, PIF reduced skin ulceration, liver inflammation and colon ulceration while maintaining beneficial anti-cancer, graft-vs.-leukemia effect. Clinical-grade PIF has high-safety profile even at supraphysiological doses. The FDA awarded Fast-Track designation, and university-sponsored clinical trials for autoimmune disorder are ongoing. Altogether, PIF properties point to its determining regulatory role in immunity, inflammation and transplant acceptance. Specific plans for using PIF for the treatment of complex neurological disorders (ie. traumatic brain injury, progressive paralysis), including neuroprotection from newborn to adult, are presented.


Assuntos
Neuroproteção/fisiologia , Peptídeos/farmacologia , Proteínas da Gravidez/metabolismo , Animais , Processos Autotróficos/fisiologia , Feminino , Doença Enxerto-Hospedeiro/tratamento farmacológico , Humanos , Recém-Nascido Prematuro/fisiologia , Inflamação/tratamento farmacológico , Doenças do Sistema Nervoso/tratamento farmacológico , Neurogênese/efeitos dos fármacos , Neurogênese/fisiologia , Gravidez , Úlcera Cutânea/tratamento farmacológico
4.
Cell Death Differ ; 22(12): 2078-86, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25976303

RESUMO

A synthetic peptide (sPIF) analogous to the mammalian embryo-derived PreImplantation Factor (PIF) enables neuroprotection in rodent models of experimental autoimmune encephalomyelitis and perinatal brain injury. The protective effects have been attributed, in part, to sPIF's ability to inhibit the biogenesis of microRNA let-7, which is released from injured cells during central nervous system (CNS) damage and induces neuronal death. Here, we uncover another novel mechanism of sPIF-mediated neuroprotection. Using a clinically relevant rat newborn brain injury model, we demonstrate that sPIF, when subcutaneously administrated, is able to reduce cell death, reverse neuronal loss and restore proper cortical architecture. We show, both in vivo and in vitro, that sPIF activates cyclic AMP dependent protein kinase (PKA) and calcium-dependent protein kinase (PKC) signaling, leading to increased phosphorylation of major neuroprotective substrates GAP-43, BAD and CREB. Phosphorylated CREB in turn facilitates expression of Gap43, Bdnf and Bcl2 known to have important roles in regulating neuronal growth, survival and remodeling. As is the case in sPIF-mediated let-7 repression, we provide evidence that sPIF-mediated PKA/PKC activation is dependent on TLR4 expression. Thus, we propose that sPIF imparts neuroprotection via multiple mechanisms at multiple levels downstream of TLR4. Given the recent FDA fast-track approval of sPIF for clinical trials, its potential clinical application for treating other CNS diseases can be envisioned.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fármacos Neuroprotetores/farmacologia , Peptídeos/farmacologia , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Modelos Animais de Doenças , Proteína GAP-43/genética , Proteína GAP-43/metabolismo , Camundongos , MicroRNAs/genética , MicroRNAs/metabolismo , Fármacos Neuroprotetores/síntese química , Peptídeos/síntese química , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Interferência de RNA , Ratos , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Proteína de Morte Celular Associada a bcl/genética , Proteína de Morte Celular Associada a bcl/metabolismo
5.
Thromb Haemost ; 112(1): 73-8, 2014 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-24499991

RESUMO

Estimating the risk of venous thromboembolism (VTE) associated with combined hormonal contraceptives following early terminated pregnancies or birth, a Danish nationwide retrospective cohort observing a one-year follow-up was defined using three unique registries. All Danish women with confirmed pregnancies aged 15-49 during the period of 1995-2009 were included. The main outcomes were relative and absolute risks of first time venous thromboembolism in users as well as non-users of combined hormonal contraceptives. In 985,569 person-years, 598 venous thromboembolisms were recorded. After early terminated pregnancies and births, respectively, 113 and 485 events occurred in 212,552 and 773,017 person-years. After early terminated pregnancies, the crude VTE incidence ratios were similar, and the numbers needed to harm were equal between groups that did or did not use combined hormonal contraceptives throughout the follow-up year. After childbirth, individuals that used combined hormonal contraceptives were more likely than non-users to experience VTE depicted by crude incidence ratios; however, the difference was only significant after 14 weeks. This implied that the numbers needed to harm were lower for those that used compared to those that did not use combined oral contraceptives in the initial 14 weeks postpartum. In conclusion, the use of combined hormonal contraceptives after early terminated pregnancies was not detrimental, but during the puerperal period, they should be used with caution.


Assuntos
Aborto Induzido/estatística & dados numéricos , Anticoncepcionais Orais Hormonais/administração & dosagem , Tromboembolia Venosa/epidemiologia , Adolescente , Adulto , Estudos de Coortes , Anticoncepcionais Orais Hormonais/efeitos adversos , Dinamarca , Feminino , Seguimentos , Humanos , Incidência , Pessoa de Meia-Idade , Período Pós-Parto , Sistema de Registros , Estudos Retrospectivos , Risco , Resultado do Tratamento , Tromboembolia Venosa/induzido quimicamente , Tromboembolia Venosa/prevenção & controle , Adulto Jovem
6.
J Thromb Haemost ; 10(7): 1320-5, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22578003

RESUMO

BACKGROUND: Inherited thrombophilias have inconsistently been linked to adverse pregnancy outcomes. Differences in study design, size and population could explain this heterogeneity. OBJECTIVE: The aim of the present study was to evaluate if factor (F)V Leiden G1691A, prothrombin mutation G20210A (PTM) and methylenetetrahydrofolate reductase C677T (MTHFR) increased the risk of severe preeclampsia, fetal growth restriction, very preterm delivery, placental abruption and a composite of these outcomes also including stillbirth. PATIENTS AND METHODS: In a nested case-cohort study of pregnant women in Denmark, we genotyped 2032 cases and 1851 random controls. Each of the medical records of the cases was validated. We calculated both genomic and allelic models, and present both models. We also performed sensitivity analyses adjusting for parity, age, smoking, body mass index and socioeconomic status. RESULTS: In the allelic models, FV Leiden increased the risk of the composite outcome (odds ratio [OR] 1.4, 95% confidence interval [CI] 1.1-1.8), severe preeclampsia (OR 1.6, 95% CI 1.1-2.4), fetal growth restriction (OR 1.4, 95% CI 1.1-1.8) and placental abruption (OR = 1.7 (95% CI 1.2-2.4). In the sensitivity analyses, adjustment diminished these estimates slightly. PTM was not significantly associated with any of the outcomes, and MTHFR was only significantly associated with severe preeclampsia (OR 1.3, 95% CI 1.1-1.6). CONCLUSION: FV Leiden predisposes to adverse pregnancy outcomes in a setting of Scandinavian women.


Assuntos
Complicações Hematológicas na Gravidez/fisiopatologia , Resultado da Gravidez , Trombofilia/complicações , Estudos de Casos e Controles , Estudos de Coortes , Dinamarca , Feminino , Humanos , Gravidez , Trombofilia/fisiopatologia
7.
J Thromb Haemost ; 8(11): 2472-7, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20626616

RESUMO

BACKGROUND: We recently described an inherited coagulopathy arising in an inbred colony of WAG/RijYcb rats. The bleeding phenotype, demonstrated by both male and female rats, included periarticular hemorrhage, spontaneous bruising, prolonged bleeding from minor wounds and maternal peripartum deaths. Coagulation testing of affected rats revealed normal prothrombin time but prolongation of activated partial thromboplastin time to twice that of controls. OBJECTIVE: To determine the specific coagulation factor and the underlying genetic defect responsible for the inherited coagulopathy in the WAG/RijYcb rats. RESULTS: Evaluation of individual clotting factor activities revealed that the affected animals had a specific deficiency of factor (F) VIII (FVIII). The FVIII gene (F8) has an autosomal location on chromosome 18 in rats, in contrast to its location on the X chromosome in mice and humans. Sequencing of F8 cDNA led to the identification of a point mutation resulting in a substitution, Leu176Pro, in the A1 domain, that is predicted to disrupt the tertiary structure of the FVIII molecule. Administration of human plasma or human recombinant FVIII corrects the coagulation abnormality in the affected animals. CONCLUSIONS: We have now identified the genetic basis of the hemostatic defect in the WAG/RijYcb rat colony. The larger size of rats relative to mice and the presence of this coagulation defect in both sexes provide a unique model, well-suited to the development of novel therapies for acquired and hereditary FVIII deficiencies.


Assuntos
Fator VIII/genética , Fator VIII/fisiologia , Hemofilia A/genética , Mutação , Alelos , Sequência de Aminoácidos , Animais , Coagulação Sanguínea , Hemostasia , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Tempo de Protrombina , Ratos , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
8.
BJOG ; 117(3): 274-81, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20015308

RESUMO

OBJECTIVE: Preterm delivery has been shown to be associated with subsequent maternal cardiovascular morbidity. However, the impact of the severity and recurrence of preterm delivery on the risk of specific cardiovascular events and the metabolic syndrome in the mother, have not been investigated. DESIGN: National registry-based retrospective cohort study. SETTING: Women delivering in Denmark from 1978 to 2007. POPULATION: Women with a first singleton delivery (n = 782 287), and with a first and second singleton delivery (n = 536 419). METHODS: Cox proportional hazard models, with the gestational age stratified into four groups as primary exposure. We made adjustments for maternal age, year of delivery, hypertensive pregnancy disorders, fetal growth deviation, placental abruption and stillbirth. MAIN OUTCOME MEASURES: Subsequent maternal hypertension, ischaemic heart diseases, thromboembolism and type-II diabetes. RESULTS: After a first delivery at 32-36 completed weeks of gestation, the adjusted risk of subsequent type-II diabetes increased 1.89-fold (1.69-2.10) and the risk of thromboembolism increased 1.42-fold (1.24-1.62). Women having a preterm delivery in the first pregnancy and a term delivery in the second had a 1.58-fold (1.34-1.86) increased risk of type-II diabetes and a 1.18-fold (0.96-1.44) increased risk of thromboembolism. Women having two preterm deliveries had a 2.30-fold (1.71-3.10) increased risk of type-II diabetes and a 1.80-fold (1.29-2.50) increased risk of thromboembolism. CONCLUSIONS: Preterm delivery is independent of other pregnancy complications associated with subsequent maternal overt type-II diabetes and thromboembolism. The recurrence of preterm delivery will augment these risks.


Assuntos
Doenças Cardiovasculares/etiologia , Diabetes Mellitus Tipo 2/etiologia , Trabalho de Parto Prematuro , Adolescente , Adulto , Métodos Epidemiológicos , Feminino , Idade Gestacional , Humanos , Hipertensão/etiologia , Pessoa de Meia-Idade , Isquemia Miocárdica/etiologia , Gravidez , Recidiva , Tromboembolia/etiologia , Adulto Jovem
9.
Thromb Res ; 124(5): 516-20, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19720393

RESUMO

Vascular injury increases access and binding of plasma-derived factor VII to perivascular cell membrane-bound tissue factor (TF). The resulting TF/VIIa complex promotes hemostasis by cleaving pro-thrombin to thrombin leading to the fibrin clot. In human pregnancy, decidual cell-expressed TF prevents decidual hemorrhage (abruption). During placentation, trophoblasts remodel decidual spiral arteries into high conductance vessels. Shallow trophoblast invasion impedes decidual vascular conversion, producing an inadequate uteroplacental blood flow that elicits abruption-related placental ischemia. Thrombin induces several biological effects via cell surface protease activated receptors. In first trimester human DCs thrombin increases synthesis of sFlt-1, which elicits placental ischemia by impeding angiogenesis-related decidual vascular remodeling. During pregnacy, the fibrillar collagen-rich amnion and choriodecidua extracellular matrix (ECM) provides greater than additive tensile strength and structural integrity. Thrombin acts as an autocrine/paracrine mediator that degrades these ECMs by augmenting decidual cell expression of: 1) matrix metalloproteinases and 2) interleukin-8, a key mediator of abruption-associated decidual infiltration of neutrophils, which express several ECM degrading proteases. Among the cell types at the maternal fetal interface at term, TF expression is highest in decidual cells indicating that this TF meets the hemostatic demands of labor and delivery. TF expression in cultured term decidual cells is enhanced by progestin and thrombin suggesting that the maintenance of elevated circulating progesterone provides hemostatic protection and that abruption-generated thrombin acts in an autocrine/paracrine fashion on decidual cells to promote hemostasis via enhanced TF expression.


Assuntos
Descolamento Prematuro da Placenta/metabolismo , Decídua/metabolismo , Complicações Hematológicas na Gravidez/metabolismo , Tromboplastina/metabolismo , Hemorragia Uterina/metabolismo , Útero/irrigação sanguínea , Descolamento Prematuro da Placenta/sangue , Descolamento Prematuro da Placenta/patologia , Animais , Decídua/patologia , Feminino , Hemostasia , Humanos , Camundongos , Camundongos Transgênicos , Gravidez , Complicações Hematológicas na Gravidez/sangue , Complicações Hematológicas na Gravidez/patologia , Trombina/metabolismo , Hemorragia Uterina/sangue , Hemorragia Uterina/patologia , Útero/metabolismo
10.
Obstet Med ; 2(1): 17-20, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27582800

RESUMO

The objective of this study was to evaluate the maternal and perinatal outcome of women with liver dysfunction during pregnancy. The study involved a prospective observational study design and was carried out at the Dow University of Health Sciences and Civil Hospital Karachi, Pakistan. A total of 800 women, who delivered during the study period from January 2006 to September 2006, constituted the study population. Pregnant women with liver dysfunction underwent evaluation for the aetiology of their liver dysfunction, including screening for hepatitis E. Thirty-five women were identified with liver dysfunction. Fourteen (40%) presented in the second trimester and 21 (60%) presented in the third trimester. Twenty-two of the 35 women (63%) had isolated acute hepatitis E; five (14%) had HELLP (haemolysis, elevated liver enzymes and low platelet count) syndrome; two (6%) had intrahepatic cholestasis of pregnancy (IHCP), two had acute fatty liver of pregnancy (AFLP) and two women had hepatitis A. A specific diagnosis was not reached in two women who died prior to delivery. In women with hepatitis E, the mean values of bilirubin and alanine transaminase were 12 mg/dL and 675 U/L, respectively. Abnormal coagulation parameters were present in 20 (57%) of the women and in 18 of 22 (82%) with hepatitis E. Fulminant hepatic failure (FHF) was seen in four patients. Seven women (20%) underwent caesarean section, 26 (74%) delivered vaginally and two women died undelivered. There were six maternal deaths in the study population; two were due to hepatitis E, one each from HELLP and AFLP, and two remained undiagnosed. The overall perinatal mortality within the group was 43%. Hepatitis E was the most common cause of FHF and maternal death in pregnant women with liver dysfunction.

11.
J Obstet Gynaecol ; 28(2): 155-60, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18393009

RESUMO

Cigarette smoking during pregnancy continues to be a significant public health concern. Maternal smoking during pregnancy has been associated with low birth weight (<2500 g), fetal growth restriction, placental problems, pre-term delivery and spontaneous abortion. Mothers who smoke during pregnancy are twice as likely to give birth to low birth weight infants, and smoking during pregnancy is estimated to be responsible for 20-30% of all low birth weight infants. Smoking during pregnancy not only affects placental function, thus causing obstetrical complications, but nicotine also crosses the placenta and acts as a neuroteratogen. This in turn, elevates the risk of cognitive and auditory processing deficits, and has also been found to be negatively associated with long-term consequences on offspring behaviour. In addition, smoking has negative long-term health consequences for both mother and child, including respiratory conditions, cancer and cardiovascular problems. This review provides insight into the genetic influences on smoking behaviour in pregnant women. In particular, the roles of genes in the neurotransmitter pathways are highlighted. It also emphasises the need for further research in this area, and provides rationale for the importance of focusing on pregnant women who are highly motivated to quit when researching smoking behaviours in women.


Assuntos
Cognição , Abandono do Hábito de Fumar , Fumar/efeitos adversos , Fumar/genética , Feminino , Humanos , Recém-Nascido de Baixo Peso , Recém-Nascido , Gravidez , Recidiva
12.
Histol Histopathol ; 22(8): 847-54, 2007 08.
Artigo em Inglês | MEDLINE | ID: mdl-17503341

RESUMO

BACKGROUND: Successful trophoblast invasion and transformation of the maternal spiral arteries requires that the pregnant endometrium (i.e., decidua) act in an immunologically paradoxical fashion, accepting the semi-allogenic placenta, while maintaining host defenses against an array of microbial pathogens. In contrast to the growing evidence that the immune surveillance molecules known as Toll-like receptors (TLRs) are expressed by trophoblasts and fetal membranes, to date, no studies have been conducted on the decidua. METHODS: Decidual tissues and cells were obtained from women undergoing first trimester elective terminations or repeat Cesarean sections and analyzed at both the protein and mRNA level. RESULTS: We now demonstrate for the first time that human decidua differentially express TLRs and their downstream signaling molecules as well as TLR stimulated induction of cytokine production in the first and third trimester of pregnancy. CONCLUSIONS: These findings suggest that the decidua is a critical component of the innate immune response in pregnancy. Moreover, the results have implications for the success or failure of compromised pregnancies in early or late gestation.


Assuntos
Decídua/metabolismo , Endométrio/irrigação sanguínea , Células Endoteliais/metabolismo , Receptores Toll-Like/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Células Cultivadas , Decídua/citologia , Decídua/efeitos dos fármacos , Decídua/imunologia , Células Endoteliais/efeitos dos fármacos , Feminino , Humanos , Imunidade Inata , Imuno-Histoquímica , Interleucinas/genética , Interleucinas/metabolismo , Lipopolissacarídeos/farmacologia , Fator 88 de Diferenciação Mieloide/metabolismo , NF-kappa B/metabolismo , Peptidoglicano/farmacologia , Poli I-C/farmacologia , Gravidez , Primeiro Trimestre da Gravidez , Terceiro Trimestre da Gravidez , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Receptores Toll-Like/genética
15.
J Thromb Haemost ; 3(3): 497-501, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15748239

RESUMO

OBJECTIVE: We posit that low levels of protein S (PS) and protein Z (PZ) contribute to adverse pregnancy outcome (APO). PATIENTS: We evaluated 103 women with subsequent normal pregnancy outcome (NPO), 106 women with APO, and 20 women with thrombophilia (TP). METHODS: We compared first trimester (1st TRI) PZ levels in 103 women with NPO, 106 women with APO, and in 20 women with TP. We compared plasma levels of PZ and free PS antigen during the second (2nd TRI) and third trimesters (3rd TRI) of pregnancy in 51 women with APO and 51 matched women with NPO. RESULTS: The mean 1st TRI PZ level was significantly lower among patients with APO, compared to pregnant controls (1.81 +/- 0.7 vs. 2.21 +/- 0.8 microg mL(-1), respectively, P < 0.001). Of patients with known TP, those with APO had a tendency for lower mean PZ levels compared to those TP women with NPO (1.5 +/- 0.6 vs. 2.3 +/- 0.9 microg mL(-1), respectively, P < 0.0631). There was a significant decrease in the PZ levels in patients with APO compared to NPO (2nd TRI 1.5 +/- 0.4 vs. 2.0 +/- 0.5 microg mL(-1), P < 0.0001; and 3rd TRI 1.6 +/- 0.5 vs. 1.9 +/- 0.5 microg mL(-1), P < 0.0002). Protein S levels were significantly lower in the 2nd and 3rd TRIs among patients with APO compared to patients with NPO (2nd TRI 34.4 +/- 11.8% vs. 38.9 +/- 10.3%, P < 0.05, respectively; and 3rd TRI 27.5 +/- 8.4 vs. 31.2 +/- 7.4, P < 0.025, respectively). CONCLUSIONS: We posit that decreased PZ and PS levels are additional risk factors for APO.


Assuntos
Proteínas Sanguíneas/análise , Complicações Hematológicas na Gravidez/sangue , Proteína S/análise , Trombofilia/sangue , Adulto , Feminino , Idade Gestacional , Humanos , Circulação Placentária , Gravidez , Resultado da Gravidez , Estudos Prospectivos , Fatores de Risco , Trombofilia/complicações
19.
Blood Coagul Fibrinolysis ; 13(3): 199-205, 2002 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11943933

RESUMO

A total of 260 consecutive patients, referred for hypercoagulable assessment, was included in this study. Four coagulation activation markers were utilized to assess these patients [enzyme-linked immunosorbent assays for soluble fibrin polymer (TpP), prothrombin fragment 1.2, thrombin-antithrombin complex, and D-dimer]. The mean levels of the activation markers directly correlated with the number of hypercoagulable abnormalities. The percentage of patients with increased TpP levels for each group was lower than the other activation markers. The findings indicate that activation markers reflect the number of underlying thrombophilic abnormalities. Our data suggest that there is a utility in performing a panel of coagulation activation markers to assess the thrombotic risk. The measurement of soluble fibrin polymer may be more reflective of an impending vascular event.


Assuntos
Produtos de Degradação da Fibrina e do Fibrinogênio/análise , Fibrina/análise , Fragmentos de Peptídeos/sangue , Peptídeo Hidrolases/sangue , Trombofilia/sangue , Resistência à Proteína C Ativada/sangue , Resistência à Proteína C Ativada/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Síndrome Antifosfolipídica/sangue , Antitrombina III , Deficiência de Antitrombina III/sangue , Doenças Autoimunes/sangue , Biomarcadores , Ensaio de Imunoadsorção Enzimática , Fator V/genética , Feminino , Humanos , Hiper-Homocisteinemia/sangue , Masculino , Pessoa de Meia-Idade , Deficiência de Proteína C/sangue , Deficiência de Proteína S/sangue , Protrombina/genética , Risco , Solubilidade , Trombofilia/etiologia , Trombofilia/genética
20.
J Allergy Clin Immunol ; 108(4 Suppl): S133-8, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11586281

RESUMO

Intravenous immune globulin (IVIG) is approved for use in a number of conditions that may occur in obstetrical patients, including autoimmune thrombocytopenia and immune deficiency syndromes. IVIG also is widely used in obstetrics for nonapproved indications, such as fetal-neonatal alloimmune thrombocytopenia, antiphospholipid syndrome, and recurrent miscarriage. This review critically analyzes the use of IVIG for these indications based on the best available information. The authors conclude IVIG is effective in the management of fetal-neonatal alloimmune thrombocytopenia. IVIG appears promising as a treatment for severe fetal-neonatal alloimmune hemolysis due to antierythrocyte antibodies. A prospective multicenter trial should be undertaken. IVIG is no more effective than heparin and low-dose aspirin in the treatment of pregnancies complicated by antiphospholipid syndrome but has not been adequately evaluated in refractory cases. Finally, pending convincing studies, IVIG is not effective and should not be used for the management of recurrent miscarriage.


Assuntos
Imunoglobulinas Intravenosas/uso terapêutico , Complicações na Gravidez/terapia , Aborto Habitual/terapia , Síndrome Antifosfolipídica/terapia , Feminino , Doenças Fetais/terapia , Humanos , Gravidez , Púrpura Trombocitopênica Idiopática/terapia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...