Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
BMC Pediatr ; 24(1): 450, 2024 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-38997672

RESUMO

BACKGROUND: Neonatal and early-life gut microbiome changes are associated with altered cardiometabolic and immune development. In this study, we explored Cesarean delivery effects on the gut microbiome in our high-risk, under-resourced Bronx, NY population. RESULTS: Fecal samples from the Bronx MomBa Health Study (Bronx MomBa Health Study) were categorized by delivery mode (vaginal/Cesarean) and analyzed via 16 S rRNA gene sequencing at four timepoints over the first two years of life. Bacteroidota organisms, which have been linked to decreased risk for obesity and type 2 diabetes, were relatively reduced by Cesarean delivery, while Firmicutes organisms were increased. Organisms belonging to the Enterococcus genus, which have been tied to aberrant immune cell development, were relatively increased in the Cesarean delivery microbiomes. CONCLUSION: Due to their far-reaching impact on cardiometabolic and immune functions, Cesarean deliveries in high-risk patient populations should be carefully considered.


Assuntos
Cesárea , Fezes , Microbioma Gastrointestinal , Humanos , Cesárea/efeitos adversos , Feminino , Recém-Nascido , Fezes/microbiologia , Cidade de Nova Iorque/epidemiologia , Gravidez , Lactente , Masculino , RNA Ribossômico 16S/genética , Firmicutes/isolamento & purificação , Enterococcus/isolamento & purificação , Bacteroidetes/isolamento & purificação
2.
Am J Hum Genet ; 103(3): 367-376, 2018 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-30173819

RESUMO

Black Americans are at increased risk for preeclampsia. Genetic variants in apolipoprotein L1 (APOL1) account for much of the increased risk for kidney disease in blacks. APOL1 is expressed in human placenta and transgenic mice expressing APOL1 develop preeclampsia. We evaluated the role of APOL1 variants in human preeclampsia. We determined maternal and fetal APOL1 genotypes in black women with preeclampsia in two populations. At Einstein Montefiore Center (EMC) Affiliated Hospitals, we studied 121 pregnancies in black women with preeclampsia. At University of Tennessee Health Science Center (UTHSC), we studied 93 pregnancies in black women with preeclampsia and 793 pregnancies without preeclampsia. We measured serum markers of preeclampsia soluble fms-like tyrosine kinase 1 (sFlt-1), placental growth factor (PlGF), and soluble endoglin (sEng). Fetal APOL1 high-risk (HR) genotype was associated with preeclampsia, with odds ratios at EMC and UTHSC of 1.84 (95% CI 1.11, 2.93) and 1.92 (95% CI 1.05, 3.49), respectively. Maternal APOL1 HR genotype was not associated with preeclampsia. Mothers with the fetal APOL1 HR genotype had more cerebral or visual disturbances (63% versus 37%, p = 0.04). In addition, fetal APOL1 HR genotype was associated with a higher sFLT-1/PlGF ratio at birth (p = 0.04). Fetal APOL1 high-risk genotype increases the risk for preeclampsia, likely by adversely affecting placental function. Further research is needed to assess whether APOL1 genetic testing can predict preeclampsia and improve pregnancy outcomes.


Assuntos
Apolipoproteína L1/genética , Biomarcadores/sangue , Negro ou Afro-Americano/genética , Feto/metabolismo , Pré-Eclâmpsia/genética , Adulto , Biomarcadores/metabolismo , Estudos de Casos e Controles , Feminino , Testes Genéticos/métodos , Genótipo , Humanos , Mães , Pré-Eclâmpsia/sangue , Pré-Eclâmpsia/metabolismo , Gravidez , Risco
3.
Am J Pathol ; 189(11): 2246-2257, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31430466

RESUMO

The mechanism by which poor maternal nutrition can affect the long-term health of offspring is poorly understood. In mice, we previously found that maternal high-fat diet (HFD) exposure results in reduced fetal growth regardless of maternal genotype. We tested our hypothesis that maternal HFD-induced inflammation contributes to metabolic disease susceptibility of the offspring via alterations in the placenta. The effect of maternal genotype, diet, and treatment with the anti-inflammatory compound N-acetylcysteine (NAC) on placental morphologic features was investigated. Placentas from wild-type dams maintained on a HFD but not those heterozygous (+/-) for Glut4 (Slc2a4) on the same diet had an increase in decidual inflammation and vasculopathy occurring together. NAC administration resulted in amelioration of HFD-induced decidual vasculopathy independent of offspring genotype and sex. Consistent with these morphologic improvements, placentas from HFD dams treated with NAC had decreased mRNA and immunostaining of IL-1ß and monocyte chemoattractant protein-1, decreased mRNA of inflammatory genes, and increased mRNA of Vegfa. These results strongly suggest consumption of an HFD results in vascular changes in placenta reflected by alterations in expression of pivotal vascular developmental markers and inflammatory genes all of which are ameliorated by NAC. These placental changes play a key role in the increased programed metabolic disease of HFD-exposed offspring.


Assuntos
Acetilcisteína/uso terapêutico , Dieta Hiperlipídica/efeitos adversos , Inflamação/prevenção & controle , Placenta/efeitos dos fármacos , Complicações na Gravidez/prevenção & controle , Doenças Vasculares/prevenção & controle , Animais , Modelos Animais de Doenças , Feminino , Inflamação/complicações , Inflamação/patologia , Masculino , Fenômenos Fisiológicos da Nutrição Materna/fisiologia , Camundongos , Camundongos Transgênicos , Placenta/patologia , Gravidez , Complicações na Gravidez/etiologia , Doenças Vasculares/complicações , Doenças Vasculares/patologia
4.
Am J Obstet Gynecol ; 220(6): 596.e1-596.e28, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30790568

RESUMO

BACKGROUND: Preterm delivery accounts for 85% of perinatal morbidity and mortality. Although the consumption of a high-fat diet leads to exaggerated proinflammatory responses and, in pregnant women, increased rates of spontaneous preterm birth, the underlying mechanisms remain unclear. OBJECTIVE: We sought to elucidate the mechanisms by which maternal consumption of a high-fat diet leads to a dysregulated immune response and, subsequently, spontaneous preterm birth. STUDY DESIGN: We performed 16S ribosomal RNA sequencing of DNA extracted and amplified from stool samples and compared the gut microbiomes of lipopolysaccharide-induced pregnant mice that were maintained on a high-fat diet compared to a normal control diet. Next, we sequenced the uterine transcriptomes of the mice. To test the effect of dampening of the immune response on the microbiome, transcriptome, and risk of spontaneous preterm birth, we induced immune tolerance with repetitive subclinical doses (0.2 mg/kg/week for 8 weeks) of endotoxin and performed 16S ribosomal RNA and uterine transcriptome sequencing on these immunotolerized mice. RESULTS: High-fat diet potentiates lipopolysaccharide-induced preterm birth by affecting the maternal gut microbiome and uterine transcriptome and reduces antioxidant capacity in a murine model. High-fat diet consumption also increases the colonization of the gut by 5 immunogenic bacteria and decreases colonization by Lachnospiraceae_NK4A136_group. Uteri from high-fat diet mice had increased expression of genes that stimulate the inflammatory-oxidative stress axis, autophagy/apoptosis, and smooth muscle contraction. Repetitive endotoxin priming protects high-fat diet dams from spontaneous preterm birth, increases colonization of the gut by Lachnospiraceae_NK4A136_group, decreases levels of immunogenic bacteria in the gut microbiome, and reduces the number of dysregulated genes after high-fat diet consumption from 994 to 74. CONCLUSION: High-fat diet-potentiated spontaneous preterm birth is mediated by increased inflammation, oxidative stress, and gut dysbiosis. The induction of immune tolerance via endotoxin priming reverses these effects and protects high-fat diet dams from spontaneous preterm birth. Based on this work, the role of immunomodulation as a novel therapeutic approach to prevent preterm birth among women who consume high-fat diets should be explored.


Assuntos
Dieta Hiperlipídica , Disbiose/imunologia , Microbioma Gastrointestinal/genética , Inflamação/genética , Estresse Oxidativo/genética , Nascimento Prematuro/imunologia , RNA Ribossômico 16S/genética , Útero/metabolismo , Animais , Apoptose/genética , Autofagia/genética , Feminino , Tolerância Imunológica , Lipopolissacarídeos , Camundongos , Contração Muscular/genética , Músculo Liso , Gravidez , Nascimento Prematuro/genética , Transcriptoma
5.
Mol Med ; 22: 747-758, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27782292

RESUMO

Previously, we have shown that N,N-dimethylacetamide (DMA) prevents inflammation-induced preterm birth in a murine model, inhibits LPS-induced increases in placental pro-inflammatory cytokines and up-regulates the anti-inflammatory cytokine Interleukin-10 (IL-10). However, DMA's mechanism of action remains to be elucidated. In the current study we investigate how DMA produces its anti-inflammatory effect. Using in vitro and ex vivo models, we show that DMA suppresses secretion of pro-inflammatory cytokines in lipopolysaccharide (LPS)-induced RAW 264.7 cells, TNFα-challenged JEG-3 cells and LPS-stimulated human placental explants. DMA significantly attenuated the secretion of TNFα, IL-6, IL-10, and granulocyte macrophage colony stimulating factor (GM-CSF) from LPS-stimulated RAW 264.7 cells, IL-6 secretion from TNFα-stimulated JEG-3 cells and TNFα, IL-6, IL-10, GM-CSF and Interleukin-8 (IL-8) from LPS-stimulated human placental explants. We further investigated if DMA's effect on cytokine expression involves the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and mitogen-activated protein kinase (MAPK) pathways. DMA (10 mM) significantly inhibited nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IκBα) degradation in LPS-stimulated RAW 264.7 cells, but there was no significant change in the expression of phosphorylated or native forms of downstream proteins in the MAPK pathway. In addition, DMA significantly attenuated luciferase activity in cells co-transfected with NF-κB-Luc reporter plasmid, but not with AP-1-Luc or CEBP-Luc reporters. Overall, our findings suggest that the anti-inflammatory activity of DMA is mediated by inhibition of the NF-κB pathway via decreased IκBα degradation.

6.
Am J Pathol ; 185(3): 862-9, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25579843

RESUMO

Premature delivery occurs in 12% of all births, and accounts for nearly half of long-term neurological morbidity, and 60% to 80% of perinatal mortality. Despite advances in obstetrics and neonatology, the rate of premature delivery has increased approximately 12% since 1990. The single most common cause of spontaneous preterm birth is infection. Several lines of evidence have demonstrated the role of endothelin-1 as both a constrictor of uterine myometrial smooth muscle and a proinflammatory mediator. Endothelin-1 activates the phospholipase C pathway, leading to activation of protein kinase C and, in turn, sphingosine kinase (SphK). The inhibition of SphK has been recently shown to control the proinflammatory response associated with sepsis. We show herein, for the first time, that SphK inhibition prevents inflammation-associated preterm birth in a murine model. Rescue of pups from premature abortion with an SphK inhibitor occurs by suppression of the proinflammatory cytokines tumor necrosis factor α, Il-1ß, and Il-6 and attenuation of polymorphonuclear inflammatory cells into the placental labyrinth. Moreover, we postulate that inhibition of SphK leads to suppression of endothelin-converting enzyme-1 expression, indicating the presence of an endothelin-converting enzyme 1/endothelin 1-SphK positive feedback loop. This work introduces a novel approach for the control of infection-triggered preterm labor, a condition for which there is no effective treatment.


Assuntos
Citocinas/metabolismo , Inibidores Enzimáticos/uso terapêutico , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Placenta/metabolismo , Nascimento Prematuro/prevenção & controle , Animais , Modelos Animais de Doenças , Inibidores Enzimáticos/farmacologia , Feminino , Inflamação/metabolismo , Lipopolissacarídeos , Camundongos , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Placenta/efeitos dos fármacos , Gravidez , Nascimento Prematuro/induzido quimicamente , Nascimento Prematuro/metabolismo
7.
Toxicol Appl Pharmacol ; 282(3): 275-84, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25230003

RESUMO

Preterm birth (PTB), defined as any delivery occurring prior to the completion of 37 weeks' gestation, currently accounts for 11-12% of all births in the United States. Maternal genito-urinary infections account for up to 40% of all PTBS and induce a pro-inflammatory state in the host. The potent vasoconstrictor Endothelin-1 (ET-1) is known to be upregulated in the setting of infection, and elicits its effect by binding to the ETA receptor. We have previously shown that antagonism of the ETA receptor with BQ-123 is capable of preventing LPS-induced PTB in mice. We hypothesize that the administration of BQ-123 post LPS exposure will dismantle a positive feedback loop observed with pro-inflammatory cytokines upstream of ET-1. On GD 15.5, pregnant C57BL/6 mice were injected with PBS, LPS, BQ-123, or LPS+BQ-123. Changes at both the level of transcription and translation were observed in uterus and placenta in the ET-1 axis and in pro- and anti-inflammatory cytokines over the course of 12h. We discovered that BQ-123, when administered 10h post LPS, is capable of increasing production of uterine and placental Interleukin-10, causing a shift away from the pro-inflammatory state. We also observed that antagonism of the ETA receptor decreased IL-1ß and TNFα in the placenta while also decreasing transcription of ET-1 in the uterus. Our results reinforce the role of ET-1 at the maternal fetal interface and highlight the potential benefit of ETA receptor blockade via the suppression of ET-1, and induction of a Th2 cytokine dominant state.


Assuntos
Citocinas/metabolismo , Antagonistas do Receptor de Endotelina A/farmacologia , Peptídeos Cíclicos/farmacologia , Placenta/efeitos dos fármacos , Complicações na Gravidez/prevenção & controle , Útero/efeitos dos fármacos , Animais , Citocinas/genética , Endotelina-1/genética , Feminino , Lipopolissacarídeos , Camundongos Endogâmicos C57BL , Placenta/metabolismo , Gravidez , Complicações na Gravidez/induzido quimicamente , Complicações na Gravidez/genética , Complicações na Gravidez/metabolismo , Útero/metabolismo
9.
Am J Pathol ; 183(2): 422-30, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23770347

RESUMO

The proinflammatory response leads to various types of pathologic pathways, including the development of preterm birth. Preterm birth occurs in 12% of deliveries in the United States and causes more than 70% of perinatal morbidity and mortality. The most common cause of spontaneous preterm birth is intrauterine infection in the mother. There is accumulating evidence indicating that the release of proinflammatory cytokines plays a critical role in the pathogenesis of inflammation-associated premature delivery. We found that the common organic solvent, N,N-dimethylacetamide (DMA), prevents endotoxin-induced preterm birth in timed pregnant C57BL/6 embryonic day (E)15.5 mice and rescues their pups from spontaneous abortion at doses many-fold lower than those currently used clinically and in a dose-dependent fashion. We also provide histologic evidence that DMA suppresses the endotoxin-triggered proinflammatory response by significantly attenuating inflammatory cell infiltration of placental tissue. Furthermore, immunoblotting analysis of placental tissue harvested from our murine models revealed DMA-mediated regulation of expression of the proinflammatory cytokines IL-1ß, tumor necrosis factor α, and IL-6, and increased expression of the regulatory inflammatory cytokine IL-10. By using in vitro studies, we provide evidence that DMA suppresses macrophage function and that this small molecule prevents nuclear translocation of nuclear factor-kB. These results suggest that DMA represents a newly discovered, nontoxic therapy for a broad range of inflammatory disorders.


Assuntos
Acetamidas/farmacologia , Anti-Inflamatórios/farmacologia , Citocinas/efeitos dos fármacos , Endotoxinas/toxicidade , Nascimento Prematuro/prevenção & controle , Animais , Feminino , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/efeitos dos fármacos , Gravidez , Nascimento Prematuro/induzido quimicamente
10.
Med Rev (2021) ; 4(3): 235-238, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38919399

RESUMO

The protein, N-myc downstream-regulated gene 2 (NDRG2), a tumor suppressor, is significantly decreased or absent in many types of cancer. There is a significant negative correlation between the levels of NDRG2 and the development and progression of cancer tumor recurrence and tumor invasion, in different cancers. In contrast, the in vitro and in vivo overexpression of the NDRG2 protein decreases the proliferation, growth, adhesion and migration of many types of cancer cells. The in vitro overexpression of NDRG2 increases the efficacy of certain anticancer drugs in specific types of cancer cells. We hypothesize that the delivery of the mRNA of the NDRG2 protein, encapsulated by lipid nanoparticles, could represent a potential treatment of metastatic and drug-resistant cancers. This would be accomplished using a self-amplifying mRNA that encodes the NDRG2 protein and an RNA-dependent-RNA polymerase, obtained from an in vitrotranscribed (IVT) mRNA. The IVT mRNA would be encapsulated in a lipid nanoformulation. The efficacy of the nanoformulation would be determined in cultured cancer cells and if the results are positive, nude mice transplanted with either drug-resistant or metastatic drug-resistant cancer cells, would be treated with the nano- formulation and monitored for efficacy and adverse effects. If the appropriate preclinical studies indicate this formulation is efficacious and safe, it is possible it could be evaluated in clinical trials.

11.
Biomolecules ; 14(4)2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38672526

RESUMO

The Biomolecules Editorial Office retracts the article, "Palmitic Acid Impedes Extravillous Trophoblast Activity by Increasing MRP1 Expression and Function" [...].

12.
Res Sq ; 2023 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-37886595

RESUMO

Alzheimer's disease (AD) is a complex neurodegenerative disease associated with memory decline, cognitive impairment, amyloid plaque formation and tau tangles. Neuroinflammation has been shown to be a precursor to apparent amyloid plaque accumulation and subsequent synaptic loss and cognitive decline. In this study, the ability of a novel, small molecule, T-ALZ01, to inhibit neuroinflammatory processes was analyzed. T-ALZ01, an inhibitor of complement component C1r, demonstrated a significant reduction in the levels of the inflammatory cytokines, IL-6 and TNF-α in vitro. An LPS-induced animal model, whereby animals were injected intraperitoneally with 0.5 mg/kg LPS, was used to analyze the effect of T-ALZ01 on neuroinflammation in vivo. Moreover, exosomes (nanosized, endogenous extracellular vehicles) were used as drug delivery vehicles to facilitate intranasal administration of T-ALZ01 across the blood-brain barrier. T-ALZ01 demonstrated significant reduction in degenerating neurons and the activation of resident microglia and astrocytes, as well as inflammatory markers in vivo. This study demonstrates a significant use of small molecule complement inhibitors via exosome drug delivery as a possible therapeutic in disorders characterized by neuroinflammation, such AD.

13.
Biomolecules ; 13(8)2023 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-37627289

RESUMO

Babies born to severe acute respiratory syndrome corona virus-2 (SARS-CoV-2)-infected mothers are at greater risk for perinatal morbidity and more likely to receive a neurodevelopmental diagnosis in the first year of life. However, the effect of maternal infection on placental function and neonatal outcomes varies depending upon the patient population. We set out to test our hypothesis that maternal SARS-CoV-2 infection in our underserved, socioeconomically disadvantaged, mostly unvaccinated, predominantly African American and Latina population in the Bronx, NY would have effects evident at birth. Under IRB approval, 56 SARS-CoV-2-positive patients infected during the "first wave" of the pandemic with alpha and beta strains of the virus, 48 patients infected during the "second wave" of the pandemic with delta and omicron strains and 61 negative third-trimester high-risk patients were randomly selected from Montefiore Medical Center (MMC), Bronx, NY. In addition, two positive cases from Yale New Haven Hospital, CT were included as controls. All 104 placentas delivered by SARS-CoV-2-positive mothers were uninfected by the virus, based on immunohistochemistry, in situ hybridization, and qPCR analysis. However, placental villous infarcts were significantly increased in first-wave cases compared to second-wave cases or negative controls. Significantly lower Apgar scores at 1 min and 5 min were observed in neonates born to infected mothers with severe symptoms. These findings suggest that even without entering the placenta, SARS-CoV-2 can affect various systemic pathways, culminating in altered placental development and function, which may adversely affect the fetus, especially in a high-risk patient population such as ours. These results underline the importance of vaccination among pregnant women, particularly in low-resource areas.


Assuntos
COVID-19 , Feminino , Humanos , Recém-Nascido , Gravidez , Índice de Apgar , COVID-19/epidemiologia , Infarto , Mães , Placenta , Gestantes , SARS-CoV-2
14.
Sci Rep ; 13(1): 7077, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-37127686

RESUMO

Alzheimer's disease (AD) is a chronic degenerative brain disorder with no clear pathogenesis or effective cure, accounting for 60-80% of cases of dementia. In recent years, the importance of neuroinflammation in the pathogenesis of AD and other neurodegenerative disorders has come into focus. Previously, we made the serendipitous discovery that the widely used drug excipient N,N-dimethylacetamide (DMA) attenuates endotoxin-induced inflammatory responses in vivo. In the current work, we investigate the effect of DMA on neuroinflammation and its mechanism of action in in-vitro and ex-vivo models of AD. We show that DMA significantly suppresses the production of inflammatory mediators, such as reactive oxygen species (ROS), nitric oxide (NO) and various cytokines and chemokines, as well as amyloid-ß (Aß), in cultured microglia and organotypic hippocampal slices induced by lipopolysaccharide (LPS). We also demonstrate that DMA inhibits Aß-induced inflammation. Finally, we show that the mechanism of DMA's effect on neuroinflammation is inhibition of the nuclear factor kappa-B (NF-κB) signaling pathway and we show how DMA dismantles the positive feedback loop between NF-κB and Aß synthesis. Taken together, our findings suggest that DMA, a generally regarded as safe compound that crosses the blood brain barrier, should be further investigated as a potential therapy for Alzheimer's disease and neuroinflammatory disorders.


Assuntos
Doença de Alzheimer , Humanos , NF-kappa B/metabolismo , Doenças Neuroinflamatórias , Transdução de Sinais , Peptídeos beta-Amiloides/metabolismo
15.
Am J Physiol Endocrinol Metab ; 302(5): E522-31, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22167521

RESUMO

Alterations in insulin signaling as well as insulin action predispose to infertility as well as adverse pregnancy outcomes; however, little is known about the role of glucagon signaling in reproduction. The glucagon receptor knockout (Gcgr(-/-)) mouse created by our laboratory was used to define the role of glucagon signaling in maintaining normal reproduction. In this mouse model, lack of glucagon signaling did not alter the hypothalamic-pituitary-ovarian axis. Pregnant Gcgr(-/-) female mice displayed persistent hypoglycemia and hyperglucagonemia. Gcgr(-/-) pregnancies were associated with decreased fetal weight, increased late-gestation fetal demise, and significant abnormalities of placentation. Gcgr(-/-) placentas contained areas of extensive mineralization, fibrinoid necrosis, narrowing of the vascular channels, and a thickened interstitium associated with trophoblast hyperplasia. Absent glucagon signaling did not alter glycogen content in Gcgr(-/-) placentas but significantly downregulated genes that control growth, adrenergic signaling, vascularization, oxidative stress, and G protein-coupled receptors. Our data suggest that, similarly to insulin, glucagon action contributes to normal female reproductive function.


Assuntos
Doenças Fetais/etiologia , Glucagon/fisiologia , Hipoglicemia/etiologia , Doenças Placentárias/etiologia , Gravidez/fisiologia , Receptores de Glucagon/fisiologia , Animais , Feminino , Morte Fetal/etiologia , Doenças Fetais/metabolismo , Retardo do Crescimento Fetal/etiologia , Regulação da Expressão Gênica no Desenvolvimento , Glucagon/sangue , Heterozigoto , Hipoglicemia/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovário/efeitos dos fármacos , Ovário/fisiologia , Adeno-Hipófise/metabolismo , Adeno-Hipófise/patologia , Placenta/metabolismo , Placenta/patologia , Doenças Placentárias/metabolismo , Doenças Placentárias/patologia , Placentação , Proteínas da Gravidez/genética , Proteínas da Gravidez/metabolismo , Receptores de Glucagon/genética , Transdução de Sinais , Superovulação/efeitos dos fármacos
16.
Pharmaceutics ; 14(10)2022 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-36297454

RESUMO

Preterm birth (PTB) is a leading cause of infant morbidity and mortality in the world. In 2020, 1 in 10 infants were born prematurely in the United States. The World Health Organization estimates that a total of 15 million infants are born prematurely every year. Current therapeutic interventions for PTB have had limited replicable success. Recent advancements in the field of nanomedicine have made it possible to utilize the vaginal administration route to effectively and locally deliver drugs to the female reproductive tract. Additionally, studies using murine models have provided important insights about the cervix as a gatekeeper for pregnancy and parturition. With these recent developments, the field of reproductive biology is on the cusp of a paradigm shift in the context of treating PTB. The present review focuses on the complexities associated with treating the condition and novel therapeutics that have produced promising results in preclinical studies.

17.
Biomolecules ; 12(8)2022 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-36009056

RESUMO

Normal function of placental extravillous trophoblasts (EVTs), which are responsible for uteroplacental vascular remodeling, is critical for adequate delivery of oxygen and nutrients to the developing fetus and normal fetal programming. Proliferation and invasion of spiral arteries by EVTs depends upon adequate levels of folate. Multidrug resistance-associated protein 1 (MRP1), which is an efflux transporter, is known to remove folate from these cells. We hypothesized that palmitic acid increases MRP1-mediated folate removal from EVTs, thereby interfering with EVTs' role in early placental vascular remodeling. HTR-8/SVneo and Swan-71 cells, first trimester human EVTs, were grown in the absence or presence of 0.5 mM and 0.7 mM palmitic acid, respectively, for 72 h. Palmitic acid increased ABCC1 gene expression and MRP1 protein expression in both cell lines. The rate of folate efflux from the cells into the media increased with a decrease in migration and invasion functions in the cultured cells. Treatment with N-acetylcysteine (NAC) prevented the palmitic acid-mediated upregulation of MRP1 and restored invasion and migration in the EVTs. Finally, in an ABCC1 knockout subline of Swan-71 cells, there was a significant increase in invasion and migration functions. The novel finding in this study that palmitic acid increases MRP1-mediated folate efflux provides a missing link that helps to explain how maternal consumption of saturated fatty acids compromises the in utero environment.


Assuntos
Placenta , Trofoblastos , Movimento Celular/fisiologia , Feminino , Ácido Fólico/metabolismo , Humanos , Proteínas Associadas à Resistência a Múltiplos Medicamentos , Ácido Palmítico/metabolismo , Ácido Palmítico/farmacologia , Placenta/metabolismo , Gravidez , Trofoblastos/metabolismo , Remodelação Vascular
18.
Fortune J Health Sci ; 5: 499-509, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-37886658

RESUMO

Inflammatory bowel disease (IBD) affects almost 7 million people worldwide and is increasing in incidence. While the precise pathogenesis of IBD remains unknown, the production of inflammatory cytokines and chemokines play a central role. We have previously found that N, N-dimethylacetamide (DMA), a widely used non-toxic drug excipient, suppresses cytokine and chemokine secretion in vitro and prevents inflammation-induced preterm birth in vivo. Using sandwich enzyme-linked immunosorbent assays (ELISAs), we tested whether DMA attenuates cytokine and chemokine secretion from LPS- or TNFα-stimulated human intestinal epithelial cells and human monocytes and HMGB1 release from RAW 264.7 cells. To test our hypothesis that the mechanism of DMA's effects in in vitro and in vivo models of IBD is inhibition of the NF-κB pathway, we used western blotting to track levels of the nuclear factor kappa B (NF-κB) inhibitory molecule I kappa B alpha (IκBα) in THP-1 human monocytes in the absence or presence of DMA. Finally, we induced colitis in C57Bl/6 mice with dextran sodium sulfate (DSS) and then tested whether i.p injections of DMA at 2.1 g/kg/day attenuates clinical and histopathologic signs of colitis. DMA attenuated cytokine and chemokine release from human intestinal epithelial cells and human monocytes and HMGB1 release from RAW 264.7 cells. Importantly, DMA prevented degradation of IκBα in THP-1 cells, thereby suggesting one mechanism for DMA's effects. Finally, we show here, for the first time, that DMA attenuates clinical and histologic features of DSS-induced colitis. Based on these data, DMA should be further explored in preclinical and clinical trials for its potential as novel drug therapy for IBD.

19.
Reprod Sci ; 29(10): 2894-2907, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35349119

RESUMO

Preterm birth accounts for the majority of perinatal mortality worldwide, and there remains no FDA-approved drug to prevent it. Recently, we discovered that the common drug excipient, N,N-dimethylacetamide (DMA), delays inflammation-induced preterm birth in mice by inhibiting NF-κB. Since we reported this finding, it has come to light that a group of widely used, structurally related aprotic solvents, including DMA, N-methyl-2-pyrrolidone (NMP) and dimethylformamide (DMF), have anti-inflammatory efficacy. We show here that DMF suppresses LPS-induced TNFα secretion from RAW 264.7 cells and IL-6 and IL-8 secretion from HTR-8 cells at concentrations that do not significantly affect cell viability. Like DMA, DMF protects IκBα from degradation and prevents the p65 subunit of NF-κB from translocating to the nucleus. In vivo, DMF decreases LPS-induced inflammatory cell infiltration and expression of TNFα and IL-6 in the placental labyrinth, all to near baseline levels. Finally, DMF decreases the rate of preterm birth in LPS-induced pregnant mice (P<.0001) and the rate at which pups are spontaneously aborted (P<.0001). In summary, DMF, a widely used solvent structurally related to DMA and NMP, delays LPS-induced preterm birth in a murine model without overt toxic effects. Re-purposing the DMA/DMF/NMP family of small molecules as anti-inflammatory drugs is a promising new approach to delaying or reducing the incidence of inflammation-induced preterm birth and potentially attenuating other inflammatory disorders as well.


Assuntos
Dimetilformamida , Nascimento Prematuro , Acetamidas , Animais , Anti-Inflamatórios/farmacologia , Dimetilformamida/efeitos adversos , Modelos Animais de Doenças , Excipientes/efeitos adversos , Feminino , Humanos , Recém-Nascido , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/prevenção & controle , Interleucina-6 , Interleucina-8 , Lipopolissacarídeos/farmacologia , Camundongos , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Placenta/metabolismo , Gravidez , Nascimento Prematuro/induzido quimicamente , Nascimento Prematuro/prevenção & controle , Solventes/efeitos adversos , Fator de Necrose Tumoral alfa
20.
Am J Pathol ; 177(4): 1929-35, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20802183

RESUMO

Preterm birth (PTB) currently accounts for 13% of all births in the United States, with the leading cause of PTB being maternal infection. Endothelin-1, an extremely potent vasoconstrictor capable of increasing myometrial smooth muscle tone, has been shown to be up-regulated in the setting of infection in pregnancy, ultimately leading to PTB. In previous work, we have shown that infection-associated PTB is controlled in our murine model by using phospharamidon, an endothelin-converting enzyme-1 inhibitor; knocking down endothelin-converting enzyme-1 mRNA; or blocking the binding of endothelin-1 to the endothelin-A (ET(A)) receptor with either BQ-123 or with HJP-272, the 6-OH compound of our series of novel synthetic (ET(A)) receptor antagonists. In the current study, we show that HJP-272, a highly selective ET(A) receptor antagonist with an IC(50) of 70.1 nmol/L, binds in a noncompetitive manner to the ET(A) receptor. Additionally, we introduce n-propyl (HJP-286) and n-butyl (HJP-278) analogs of HJP-272. We find that the LD(50) of HJP-272, the analog in the series most effective in controlling preterm birth, is more than 20-fold higher than its therapeutic dose. Acute exposure to high doses of these compounds produces no histological changes in any organ, while chronic exposure produces only a rare hepatotoxic effect. These findings may be of clinical significance, as there is currently no FDA-approved therapy for women presenting with threatened preterm delivery.


Assuntos
Benzodioxóis/uso terapêutico , Antagonistas do Receptor de Endotelina A , Endotelina-1/antagonistas & inibidores , Hidroxiquinolinas/uso terapêutico , Complicações Infecciosas na Gravidez/prevenção & controle , Nascimento Prematuro/etiologia , Nascimento Prematuro/prevenção & controle , Quinolonas/uso terapêutico , Animais , Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ácido Aspártico Endopeptidases/metabolismo , Ligação Competitiva , Endotelina-1/metabolismo , Enzimas Conversoras de Endotelina , Feminino , Lipopolissacarídeos/farmacologia , Metaloendopeptidases/antagonistas & inibidores , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Prenhez , Receptor de Endotelina A/metabolismo , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA