Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Cell ; 184(13): 3356-3357, 2021 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-34171317

RESUMO

Education of the human immune system begins in utero via T cell activation and memory development. However, whether part of the education is provided by exposure to microbes in utero remains controversial and unclear. In this issue of Cell, Mishra et al. provide new evidence that the fetal gut may be colonized by bacteria that prime T cell memories.


Assuntos
Bactérias , Ativação Linfocitária , Feto , Humanos , Linfócitos T/imunologia
3.
Cell ; 170(2): 273-283.e12, 2017 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-28708997

RESUMO

The emergence of Zika virus (ZIKV) and its association with congenital malformations has prompted the rapid development of vaccines. Although efficacy with multiple viral vaccine platforms has been established in animals, no study has addressed protection during pregnancy. We tested in mice two vaccine platforms, a lipid nanoparticle-encapsulated modified mRNA vaccine encoding ZIKV prM and E genes and a live-attenuated ZIKV strain encoding an NS1 protein without glycosylation, for their ability to protect against transmission to the fetus. Vaccinated dams challenged with a heterologous ZIKV strain at embryo day 6 (E6) and evaluated at E13 showed markedly diminished levels of viral RNA in maternal, placental, and fetal tissues, which resulted in protection against placental damage and fetal demise. As modified mRNA and live-attenuated vaccine platforms can restrict in utero transmission of ZIKV in mice, their further development in humans to prevent congenital ZIKV syndrome is warranted.


Assuntos
Vacinas Virais/administração & dosagem , Infecção por Zika virus/imunologia , Infecção por Zika virus/prevenção & controle , Zika virus/fisiologia , Aedes/virologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Células Sanguíneas/virologia , Embrião de Mamíferos/virologia , Feminino , Feto/virologia , Humanos , Lipídeos/administração & dosagem , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Organismos Livres de Patógenos Específicos , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/imunologia , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/imunologia , Vacinas Virais/imunologia , Infecção por Zika virus/virologia
4.
Cell ; 165(5): 1081-1091, 2016 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-27180225

RESUMO

Zika virus (ZIKV) infection in pregnant women causes intrauterine growth restriction, spontaneous abortion, and microcephaly. Here, we describe two mouse models of placental and fetal disease associated with in utero transmission of ZIKV. Female mice lacking type I interferon signaling (Ifnar1(-/-)) crossed to wild-type (WT) males produced heterozygous fetuses resembling the immune status of human fetuses. Maternal inoculation at embryonic day 6.5 (E6.5) or E7.5 resulted in fetal demise that was associated with ZIKV infection of the placenta and fetal brain. We identified ZIKV within trophoblasts of the maternal and fetal placenta, consistent with a trans-placental infection route. Antibody blockade of Ifnar1 signaling in WT pregnant mice enhanced ZIKV trans-placental infection although it did not result in fetal death. These models will facilitate the study of ZIKV pathogenesis, in utero transmission, and testing of therapies and vaccines to prevent congenital malformations.


Assuntos
Modelos Animais de Doenças , Doenças Fetais/virologia , Doenças Placentárias/virologia , Complicações Infecciosas na Gravidez/virologia , Infecção por Zika virus/patologia , Zika virus/fisiologia , Animais , Apoptose , Encéfalo/embriologia , Encéfalo/patologia , Encéfalo/virologia , Feminino , Doenças Fetais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Doenças Placentárias/patologia , Gravidez , Complicações Infecciosas na Gravidez/patologia , RNA Viral/isolamento & purificação , Receptor de Interferon alfa e beta/genética , Receptor de Interferon alfa e beta/metabolismo , Infecção por Zika virus/virologia
5.
Nat Immunol ; 18(11): 1261-1269, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28945244

RESUMO

The Zika virus (ZIKV) epidemic has resulted in congenital abnormalities in fetuses and neonates. Although some cross-reactive dengue virus (DENV)-specific antibodies can enhance ZIKV infection in mice, those recognizing the DENV E-dimer epitope (EDE) can neutralize ZIKV infection in cell culture. We evaluated the therapeutic activity of human monoclonal antibodies to DENV EDE for their ability to control ZIKV infection in the brains, testes, placentas, and fetuses of mice. A single dose of the EDE1-B10 antibody given 3 d after ZIKV infection protected against lethality, reduced ZIKV levels in brains and testes, and preserved sperm counts. In pregnant mice, wild-type or engineered LALA variants of EDE1-B10, which cannot engage Fcg receptors, diminished ZIKV burden in maternal and fetal tissues, and protected against fetal demise. Because neutralizing antibodies to EDE have therapeutic potential against ZIKV, in addition to their established inhibitory effects against DENV, it may be possible to develop therapies that control disease caused by both viruses.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Antivirais/imunologia , Vírus da Dengue/imunologia , Epitopos/imunologia , Proteínas do Envelope Viral/imunologia , Infecção por Zika virus/imunologia , Animais , Encéfalo/imunologia , Encéfalo/virologia , Chlorocebus aethiops , Reações Cruzadas/imunologia , Vírus da Dengue/classificação , Vírus da Dengue/metabolismo , Feminino , Feto/imunologia , Feto/virologia , Interações Hospedeiro-Patógeno/imunologia , Humanos , Masculino , Camundongos , Testes de Neutralização , Gravidez , Multimerização Proteica/imunologia , Testículo/imunologia , Testículo/virologia , Células Vero , Proteínas do Envelope Viral/química , Carga Viral/imunologia , Zika virus/imunologia , Zika virus/fisiologia , Infecção por Zika virus/virologia
6.
Dev Biol ; 493: 29-39, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36368522

RESUMO

A global increase in older individuals creates an increasing demand to understand numerous healthcare challenges related to aging. This population is subject to changes in tissue physiology and the immune response network. Older individuals are particularly susceptible to infectious diseases, with one of the most common being urinary tract infections (UTIs). Postmenopausal and older women have the highest risk of recurrent UTIs (rUTIs); however, why rUTIs become more frequent after menopause and during old age is incompletely understood. This increased susceptibility and severity among older individuals may involve functional changes to the immune system with age. Aging also has substantial effects on the epithelium and the immune system that led to impaired protection against pathogens, yet heightened and prolonged inflammation. How the immune system and its responses to infection changes within the bladder mucosa during aging has largely remained poorly understood. In this review, we highlight our understanding of bladder innate and adaptive immunity and the impact of aging and hormones and hormone therapy on bladder epithelial homeostasis and immunity. In particular, we elaborate on how the cellular and molecular immune landscape within the bladder can be altered during aging as aged mice develop bladder tertiary lymphoid tissues (bTLT), which are absent in young mice leading to profound age-associated change to the immune landscape in bladders that might drive the significant increase in UTI susceptibility. Knowledge of host factors that prevent or promote infection can lead to targeted treatment and prevention regimens. This review also identifies unique host factors to consider in the older, female host for improving rUTI treatment and prevention by dissecting the age-associated alteration of the bladder mucosal immune system.


Assuntos
Infecções Urinárias , Sistema Urinário , Feminino , Camundongos , Animais , Bexiga Urinária , Envelhecimento , Homeostase , Imunidade Inata
7.
J Urol ; 209(5): 928-936, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36715657

RESUMO

PURPOSE: We identify correlates and clinical outcomes of cystitis cystica, a poorly understood chronic inflammatory bladder change, in women with recurrent urinary tract infections. MATERIALS AND METHODS: A retrospective, observational cohort of women with recurrent urinary tract infections who underwent cystoscopy (n=138) from 2015 to 2018 were identified using electronic medical records. Cystitis cystica status was abstracted from cystoscopy reports and correlations were identified by logistic regression. Urinary tract infection-free survival time associated with cystitis cystica was evaluated by Cox proportional hazards regression. Exact logistic regression was used to identify factors associated with changes to cystitis cystica lesions on repeat cystoscopy. Biopsies of cystitis cystica lesions were examined by routine histology and immunofluorescence. RESULTS: Fifty-three patients (38%) had cystitis cystica on cystoscopy. Cystitis cystica was associated with postmenopausal status (OR: 5.53, 95% CI: 1.39-37.21), pelvic floor myofascial pain (6.82, 1.78-45.04), having ≥4 urinary tract infections in the past year (2.28, 1.04-5.09), and a shorter time to next urinary tract infection (HR: 1.54, 95% CI: 1.01-2.35). Forty-two patients (82%) demonstrated improvement or resolution of lesions. Ten/11 (91%) biopsied cystitis cystica lesions were tertiary lymphoid tissue with germinal centers and resembled follicular cystitis. CONCLUSIONS: Cystitis cystica lesions were associated with postmenopausal status, pelvic floor myofascial pain, and number of urinary tract infections in the prior year and predicted worse recurrent urinary tract infection outcomes. Cystitis cystica lesions are tertiary lymphoid tissue/follicular cystitis that may improve or resolve over time with treatment. Identifying cystitis cystica in recurrent urinary tract infection patients may be useful in informing future urinary tract infection risk and tailoring appropriate treatment strategies.


Assuntos
Cistite , Infecções Urinárias , Feminino , Humanos , Cistite/complicações , Cistite/tratamento farmacológico , Cistite/patologia , Tecido Linfoide/patologia , Dor/patologia , Pós-Menopausa , Estudos Retrospectivos , Bexiga Urinária/patologia , Infecções Urinárias/etiologia , Infecções Urinárias/complicações
8.
Nature ; 540(7633): 443-447, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27819683

RESUMO

Zika virus (ZIKV) is an emerging mosquito-transmitted flavivirus that can cause severe disease, including congenital birth defects during pregnancy. To develop candidate therapeutic agents against ZIKV, we isolated a panel of human monoclonal antibodies from subjects that were previously infected with ZIKV. We show that a subset of antibodies recognize diverse epitopes on the envelope (E) protein and exhibit potent neutralizing activity. One of the most inhibitory antibodies, ZIKV-117, broadly neutralized infection of ZIKV strains corresponding to African and Asian-American lineages. Epitope mapping studies revealed that ZIKV-117 recognized a unique quaternary epitope on the E protein dimer-dimer interface. We evaluated the therapeutic efficacy of ZIKV-117 in pregnant and non-pregnant mice. Monoclonal antibody treatment markedly reduced tissue pathology, placental and fetal infection, and mortality in mice. Thus, neutralizing human antibodies can protect against maternal-fetal transmission, infection and disease, and reveal important determinants for structure-based rational vaccine design efforts.


Assuntos
Anticorpos Neutralizantes/imunologia , Doenças Fetais/prevenção & controle , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Replicação Viral/imunologia , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia , Zika virus/crescimento & desenvolvimento , Zika virus/imunologia , África , América , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Neutralizantes/química , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/química , Anticorpos Antivirais/imunologia , Anticorpos Antivirais/uso terapêutico , Especificidade de Anticorpos , Ásia , Linfócitos B/imunologia , Modelos Animais de Doenças , Mapeamento de Epitopos , Feminino , Doenças Fetais/imunologia , Doenças Fetais/virologia , Feto/imunologia , Feto/virologia , Humanos , Masculino , Camundongos , Modelos Moleculares , Placenta/imunologia , Placenta/virologia , Gravidez , Multimerização Proteica , Taxa de Sobrevida , Proteínas Virais/química , Proteínas Virais/imunologia , Vacinas Virais/química , Vacinas Virais/imunologia , Infecção por Zika virus/patologia
9.
Fetal Pediatr Pathol ; 41(3): 403-412, 2022 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33040615

RESUMO

Background: This study aims to investigate whether maternal SARS-CoV-2 status affects placental pathology. Methods: A retrospective case-control study was conducted by reviewing charts and slides of placentas delivered between April 1 to July 24, 2020. Clinical history of "COVID-19" was searched in Pathology Database (CoPath). Controls were matched with SARS-CoV-2-negative women with singleton deliveries in the 3rd-trimester. Pathological features were extracted from placental pathology reports. Results: Twenty-one 3rd trimester placentas from SARS-CoV-2-positive women were identified and compared to 20 placentas from SARS-CoV-2-negative women. There were no significant differences in individual or group gross or microscopic pathological features. Within the SARS-CoV-2+ group, there are no differences between symptomatic and asymptomatic women. Conclusion: Placentas from SARS-CoV-2-positive women do not demonstrate a specific pathological pattern. Pregnancy complicated with COVID-19 during the 3rd trimester does not have a demonstrable effect on placental structure and pathology.


Assuntos
COVID-19 , Complicações Infecciosas na Gravidez , Estudos de Casos e Controles , Feminino , Humanos , Placenta/patologia , Gravidez , Complicações Infecciosas na Gravidez/patologia , Terceiro Trimestre da Gravidez , Estudos Retrospectivos , SARS-CoV-2
10.
Am J Obstet Gynecol ; 224(5): 530.e1-530.e17, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33248136

RESUMO

BACKGROUND: Group B Streptococcus is a common vaginal bacterium and the leading cause of invasive fetoplacental infections. Group B Streptococcus in the vagina can invade through the cervix to cause ascending uteroplacental infections or can be transmitted to the neonate during vaginal delivery. Some studies have found that women with a "dysbiotic" polymicrobial or Lactobacillus-depleted vaginal microbiota are more likely to harbor group B Streptococcus. Gardnerella vaginalis is often the most abundant bacteria in the vaginas of women with dysbiosis, while being detected at lower levels in most other women, and has been linked with several adverse pregnancy outcomes. Mouse models of group B Streptococcus and Gardnerella vaginalis colonization have been reported but, to the best of our knowledge, the two have not been studied together. The overarching idea driving this study is that certain members of the dysbiotic vaginal microbiota, such as Gardnerella vaginalis, may directly contribute to the increased rate of group B Streptococcus vaginal colonization observed in women with vaginal dysbiosis. OBJECTIVE: We used a mouse model to test the hypothesis that vaginal exposure to Gardnerella vaginalis may facilitate colonization and/or invasive infection of the upper reproductive tract by group B Streptococcus during pregnancy. STUDY DESIGN: Timed-pregnant mice were generated using an allogeneic mating strategy with BALB/c males and C57Bl/6 females. Dams were vaginally inoculated at gestational day 14 with group B Streptococcus alone (using a 10-fold lower dose than previously reported models) or coinoculated with group B Streptococcus and Gardnerella vaginalis. Bacterial titers were enumerated in vaginal, uterine horn, and placental tissues at gestational day 17. The presence (Fisher exact tests) and levels (Mann-Whitney U tests) of bacterial titers were compared between mono- and coinoculated dams in each compartment. Relative risks were calculated for outcomes that occurred in both groups. Tissue samples were also examined for evidence of pathophysiology. RESULTS: Inoculation of pregnant mice with 107 group B Streptococcus alone did not result in vaginal colonization or ascending infection. In contrast, coinoculation of group B Streptococcus with Gardnerella vaginalis in pregnant mice resulted in a 10-fold higher risk of group B Streptococcus vaginal colonization (relative risk, 10.31; 95% confidence interval, 2.710-59.04; P=.0006 [Fisher exact test]). Ascending group B Streptococcus infection of the uterus and placenta occurred in approximately 40% of coinoculated animals, whereas none of those receiving group B Streptococcus alone developed uterine or placental infections. Immunofluorescence microscopy revealed group B Streptococcus in both the maternal and fetal sides of the placenta. Histologic inflammation and increased proinflammatory cytokines were evident in the setting of group B Streptococcus placental infection. Interestingly, placentas from dams exposed to group B Streptococcus and Gardnerella vaginalis, but without recoverable vaginal or placental bacteria, displayed distinct histopathologic features and cytokine signatures. CONCLUSION: These data suggest that Gardnerella vaginalis vaginal exposure can promote group B Streptococcus vaginal colonization, resulting in a greater likelihood of invasive perinatal group B Streptococcus infections. These findings suggest that future clinical studies should examine whether the presence of Gardnerella vaginalis is a risk factor for group B Streptococcus vaginal colonization in women. Because Gardnerella vaginalis can also be present in women without bacterial vaginosis, these findings may be relevant both inside and outside of the context of vaginal dysbiosis.


Assuntos
Coinfecção/complicações , Gardnerella vaginalis , Doenças Placentárias/microbiologia , Infecções Estreptocócicas/microbiologia , Streptococcus agalactiae , Doenças Uterinas/microbiologia , Vaginose Bacteriana/microbiologia , Animais , Citocinas/metabolismo , Disbiose/microbiologia , Feminino , Camundongos , Interações Microbianas , Microbiota , Placenta/microbiologia , Doenças Placentárias/metabolismo , Doenças Placentárias/patologia , Gravidez , Vagina/microbiologia
11.
J Biol Chem ; 293(16): 6022-6038, 2018 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-29496999

RESUMO

Germline-encoded receptors recognizing common pathogen-associated molecular patterns are a central element of the innate immune system and play an important role in shaping the host response to infection. Many of the innate immune molecules central to these signaling pathways are evolutionarily conserved. LysMD3 is a novel molecule containing a putative peptidoglycan-binding domain that has orthologs in humans, mice, zebrafish, flies, and worms. We found that the lysin motif (LysM) of LysMD3 is likely related to a previously described peptidoglycan-binding LysM found in bacteria. Mouse LysMD3 is a type II integral membrane protein that co-localizes with GM130+ structures, consistent with localization to the Golgi apparatus. We describe here two lines of mLysMD3-deficient mice for in vivo characterization of mLysMD3 function. We found that mLysMD3-deficient mice were born at Mendelian ratios and had no obvious pathological abnormalities. They also exhibited no obvious immune response deficiencies in a number of models of infection and inflammation. mLysMD3-deficient mice exhibited no signs of intestinal dysbiosis by 16S analysis or alterations in intestinal gene expression by RNA sequencing. We conclude that mLysMD3 contains a LysM with cytoplasmic orientation, but we were unable to define a physiological role for the molecule in vivo.


Assuntos
Deleção de Genes , Animais , Autoantígenos/análise , Infecções Bacterianas/genética , Infecções Bacterianas/imunologia , Sistemas CRISPR-Cas , Feminino , Imunidade Inata , Inflamação/genética , Inflamação/imunologia , Masculino , Proteínas de Membrana/análise , Camundongos , Micoses/genética , Micoses/imunologia , Filogenia , Viroses/genética , Viroses/imunologia
12.
Am J Physiol Renal Physiol ; 316(5): F814-F822, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30724105

RESUMO

Iron is a critical nutrient required by hosts and pathogens. Uropathogenic Escherichia coli (UPEC), the principal causative agent of urinary tract infections (UTIs), chelate iron for their survival and persistence. Here, we demonstrate that dietary modulation of iron availability limits UPEC burden in a mouse model of UTI. Mice on a low-iron diet exhibit reduced systemic and bladder mucosal iron availability and harbor significantly lower bacterial burden, concomitant with dampened inflammation. Hepcidin is a master regulator of iron that controls iron-dependent UPEC intracellular growth. Hepcidin-deficient mice ( Hamp1-/-) exhibit accumulation of iron deposits, persistent bacterial burden in the bladder, and a heightened inflammatory response to UTI. However, a low-iron dietary regimen reversed the iron overload and increased bacterial burden phenotypes in Hamp1-/- mice. Thus modulation of iron levels via diet can reduce UPEC infection and persistence, which may have significant implications for clinical management of UTI.


Assuntos
Infecções por Escherichia coli/dietoterapia , Ferro da Dieta/metabolismo , Bexiga Urinária/microbiologia , Infecções Urinárias/dietoterapia , Escherichia coli Uropatogênica/patogenicidade , Animais , Carga Bacteriana , Modelos Animais de Doenças , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/microbiologia , Ferritinas/metabolismo , Hepcidinas/genética , Hepcidinas/metabolismo , Interações Hospedeiro-Patógeno , Interleucina-6/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Bexiga Urinária/metabolismo , Infecções Urinárias/metabolismo , Infecções Urinárias/microbiologia
13.
Hum Reprod ; 34(6): 1106-1116, 2019 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-31037294

RESUMO

STUDY QUESTION: Does altering gut microbiota with antibiotic treatment have any impact on endometriosis progression? SUMMARY ANSWER: Antibiotic therapy reduces endometriosis progression in mice, possibly by reducing specific gut bacteria. WHAT IS KNOWN ALREADY: Endometriosis, a chronic condition causing abdominal pain and infertility, afflicts up to 10% of women between the ages of 25 and 40, ~5 million women in the USA. Current treatment strategies, including hormone therapy and surgery, have significant side effects and do not prevent recurrences. We have little understanding of why some women develop endometriosis and others do not. STUDY DESIGN, SIZE, DURATION: Mice were treated with broad-spectrum antibiotics or metronidazole, subjected to surgically-induced endometriosis and assayed after 21 days. PARTICIPANTS/MATERIALS, SETTING, METHODS: The volumes and weights of endometriotic lesions and histological signatures were analysed. Proliferation and inflammation in lesions were assessed by counting cells that were positive for the proliferation marker Ki-67 and the macrophage marker Iba1, respectively. Differences in faecal bacterial composition were assessed in mice with and without endometriosis, and faecal microbiota transfer studies were performed. MAIN RESULTS AND THE ROLE OF CHANCE: In mice treated with broad-spectrum antibiotics (vancomycin, neomycin, metronidazole and ampicillin), endometriotic lesions were significantly smaller (~ 5-fold; P < 0.01) with fewer proliferating cells (P < 0.001) than those in mice treated with vehicle. Additionally, inflammatory responses, as measured by the macrophage marker Iba1 in lesions and IL-1ß, TNF-α, IL-6 and TGF-ß1 in peritoneal fluid, were significantly reduced in mice treated with broad-spectrum antibiotics (P < 0.05). In mice treated with metronidazole only, but not in those treated with neomycin, ectopic lesions were significantly (P < 0.001) smaller in volume than those from vehicle-treated mice. Finally, oral gavage of faeces from mice with endometriosis restored the endometriotic lesion growth and inflammation (P < 0.05 and P < 0.01, respectively) in metronidazole-treated mice. LARGE-SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: These findings are from a mouse model of surgically-induced endometriosis. Further studies are needed to determine the mechanism by which gut bacteria promote inflammation, identify bacterial genera or species that promote disease progression and assess the translatability of these findings to humans. WIDER IMPLICATIONS OF THE FINDINGS: Our findings suggest that gut bacteria promote endometriosis progression in mice. This finding if translated to humans, could aid in the development of improved diagnostic tools and personalised treatment strategies. STUDY FUNDING AND COMPETING INTEREST(S): This work was funded, in part, by: a National Institutes of Health (NIH)/ National Institute of Child Health and Human Development (NICHD) grant (R00HD080742) to RK; Washington University School of Medicine start-up funds to RK; an Endometriosis Foundation of America Research Award to R.K.; and an NIH/NICHD grant (R01HD091218) to IUM. The authors report no conflict of interest.


Assuntos
Antibacterianos/administração & dosagem , Endometriose/tratamento farmacológico , Microbioma Gastrointestinal/efeitos dos fármacos , Metronidazol/administração & dosagem , Doenças Peritoneais/tratamento farmacológico , Animais , Modelos Animais de Doenças , Progressão da Doença , Endometriose/microbiologia , Endometriose/patologia , Endométrio/patologia , Transplante de Microbiota Fecal/efeitos adversos , Fezes/microbiologia , Feminino , Microbioma Gastrointestinal/fisiologia , Humanos , Camundongos , Doenças Peritoneais/microbiologia , Doenças Peritoneais/patologia
14.
Am J Obstet Gynecol ; 221(6): 549-562, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31207234

RESUMO

The impact of infections and inflammation during pregnancy on the developing fetal brain remains incompletely defined, with important clinical and research gaps. Although the classic infectious TORCH pathogens (ie, Toxoplasma gondii, rubella virus, cytomegalovirus [CMV], herpes simplex virus) are known to be directly teratogenic, emerging evidence suggests that these infections represent the most extreme end of a much larger spectrum of injury. We present the accumulating evidence that prenatal exposure to a wide variety of viral and bacterial infections-or simply inflammation-may subtly alter fetal brain development, leading to neuropsychiatric consequences for the child later in life. The link between influenza infections in pregnant women and an increased risk for development of schizophrenia in their children was first described more than 30 years ago. Since then, evidence suggests that a range of infections during pregnancy may also increase risk for autism spectrum disorder and depression in the child. Subsequent studies in animal models demonstrated that both pregnancy infections and inflammation can result in direct injury to neurons and neural progenitor cells or indirect injury through activation of microglia and astrocytes, which can trigger cytokine production and oxidative stress. Infectious exposures can also alter placental serotonin production, which can perturb neurotransmitter signaling in the developing brain. Clinically, detection of these subtle injuries to the fetal brain is difficult. As the neuropsychiatric impact of perinatal infections or inflammation may not be known for decades after birth, our construct for defining teratogenic infections in pregnancy (eg, TORCH) based on congenital anomalies is insufficient to capture the full adverse impact on the child. We discuss the clinical implications of this body of evidence and how we might place greater emphasis on prevention of prenatal infections. For example, increasing uptake of the seasonal influenza vaccine is a key strategy to reduce perinatal infections and the risk for fetal brain injury. An important research gap exists in understanding how antibiotic therapy during pregnancy affects the fetal inflammatory load and how to avoid inflammation-mediated injury to the fetal brain. In summary, we discuss the current evidence and mechanisms linking infections and inflammation with the increased lifelong risk of neuropsychiatric disorders in the child, and how we might improve prenatal care to protect the fetal brain.


Assuntos
Inflamação/epidemiologia , Transtornos Mentais/epidemiologia , Complicações Infecciosas na Gravidez/epidemiologia , Efeitos Tardios da Exposição Pré-Natal/epidemiologia , Animais , Astrócitos/imunologia , Transtorno do Espectro Autista/epidemiologia , Transtorno do Espectro Autista/imunologia , Transtorno do Espectro Autista/metabolismo , Transtorno Bipolar/epidemiologia , Transtorno Bipolar/imunologia , Transtorno Bipolar/metabolismo , Citocinas/imunologia , Transtorno Depressivo/epidemiologia , Transtorno Depressivo/imunologia , Transtorno Depressivo/metabolismo , Feminino , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Transtornos Mentais/imunologia , Transtornos Mentais/metabolismo , Microglia/imunologia , Células-Tronco Neurais , Neurônios , Estresse Oxidativo/imunologia , Placenta/metabolismo , Gravidez , Complicações na Gravidez/epidemiologia , Complicações na Gravidez/imunologia , Complicações na Gravidez/metabolismo , Complicações Infecciosas na Gravidez/imunologia , Complicações Infecciosas na Gravidez/metabolismo , Efeitos Tardios da Exposição Pré-Natal/imunologia , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Esquizofrenia/epidemiologia , Esquizofrenia/imunologia , Esquizofrenia/metabolismo , Serotonina/metabolismo
16.
J Urol ; 209(5): 949, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36815426
17.
Dev Dyn ; 246(4): 336-343, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28109014

RESUMO

Homeostatic maintenance and repair of the urothelium upon injury are required for a functional bladder in both healthy and disease conditions. Understanding the cellular and molecular mechanisms underlying the urothelial regenerative response is key to designing strategies for tissue repair and ultimately treatments for urologic diseases including urinary tract infections, voiding dysfunction, painful bladder syndrome, and bladder cancer. In this article, we review studies on urothelial ontogeny during development and regeneration following various injury modalities. Signaling pathways involved in urothelial regeneration and in urothelial carcinogenesis are also discussed. Developmental Dynamics 246:336-343, 2017. © 2016 Wiley Periodicals, Inc.


Assuntos
Regeneração , Neoplasias Urológicas/patologia , Urotélio/crescimento & desenvolvimento , Animais , Humanos , Transdução de Sinais , Bexiga Urinária/fisiologia , Doenças Urológicas/fisiopatologia , Urotélio/lesões , Urotélio/fisiologia
18.
Nature ; 472(7341): 110-4, 2011 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-21389986

RESUMO

Epithelial integrity in metazoan organs is maintained through the regulated proliferation and differentiation of organ-specific stem and progenitor cells. Although the epithelia of organs such as the intestine regenerate constantly and thus remain continuously proliferative, other organs, such as the mammalian urinary bladder, shift from near-quiescence to a highly proliferative state in response to epithelial injury. The cellular and molecular mechanisms underlying this injury-induced mode of regenerative response are poorly defined. Here we show in mice that the proliferative response to bacterial infection or chemical injury within the bladder is regulated by signal feedback between basal cells of the urothelium and the stromal cells that underlie them. We demonstrate that these basal cells include stem cells capable of regenerating all cell types within the urothelium, and are marked by expression of the secreted protein signal Sonic hedgehog (Shh). On injury, Shh expression in these basal cells increases and elicits increased stromal expression of Wnt protein signals, which in turn stimulate the proliferation of both urothelial and stromal cells. The heightened activity of this signal feedback circuit and the associated increase in cell proliferation appear to be required for restoration of urothelial function and, in the case of bacterial injury, may help clear and prevent further spread of infection. Our findings provide a conceptual framework for injury-induced epithelial regeneration in endodermal organs, and may provide a basis for understanding the roles of signalling pathways in cancer growth and metastasis.


Assuntos
Células Epiteliais/citologia , Proteínas Hedgehog/metabolismo , Regeneração/fisiologia , Células-Tronco/citologia , Bexiga Urinária/citologia , Proteínas Wnt/metabolismo , Animais , Linhagem da Célula , Proliferação de Células , Células Epiteliais/metabolismo , Retroalimentação Fisiológica , Feminino , Fatores de Crescimento de Fibroblastos/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Organoides/citologia , Transdução de Sinais , Células-Tronco/metabolismo , Células Estromais/citologia , Células Estromais/metabolismo , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/lesões , Bexiga Urinária/metabolismo , Doenças da Bexiga Urinária/induzido quimicamente , Doenças da Bexiga Urinária/metabolismo , Doenças da Bexiga Urinária/microbiologia , Doenças da Bexiga Urinária/patologia , Escherichia coli Uropatogênica/fisiologia , Urotélio/citologia , Proteína GLI1 em Dedos de Zinco
20.
Mo Med ; 114(3): 168-170, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-30228574

RESUMO

Zika virus (ZIKV) has been linked to intrauterine growth restriction (IUGR), spontaneous miscarriage, and microcephaly in infants of women infected during pregnancy. To determine how ZIKV affects the fetus, we infected pregnant mice subcutaneously (mimicking a mosquito bite) with ZIKV. Multiple techniques revealed that ZIKV replicated within placental trophoblasts, fetal endothelial cells, and the fetal neocortex. We also noted severe placental defects, IUGR, and fetal death. Thus, our mouse model recapitulated ZIKV infection in human pregnancy and demonstrated that ZIKV can be transmitted from mother to fetus via the placenta.


Assuntos
Aborto Espontâneo/virologia , Retardo do Crescimento Fetal/virologia , Microcefalia/virologia , Placenta/virologia , Infecção por Zika virus/transmissão , Aborto Espontâneo/epidemiologia , Animais , Feminino , Morte Fetal/etiologia , Retardo do Crescimento Fetal/epidemiologia , Feto , Humanos , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Camundongos , Microcefalia/epidemiologia , Modelos Animais , Gravidez , Complicações Infecciosas na Gravidez/epidemiologia , Trofoblastos/virologia , Organização Mundial da Saúde/organização & administração , Zika virus/genética , Zika virus/isolamento & purificação , Infecção por Zika virus/complicações
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa