Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
1.
Cell ; 184(13): 3356-3357, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34171317

ABSTRACT

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.


Subject(s)
Bacteria , Lymphocyte Activation , Fetus , Humans , T-Lymphocytes/immunology
3.
Cell ; 170(2): 273-283.e12, 2017 Jul 13.
Article in English | MEDLINE | ID: mdl-28708997

ABSTRACT

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.


Subject(s)
Viral Vaccines/administration & dosage , Zika Virus Infection/immunology , Zika Virus Infection/prevention & control , Zika Virus/physiology , Aedes/virology , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Blood Cells/virology , Embryo, Mammalian/virology , Female , Fetus/virology , Humans , Lipids/administration & dosage , Male , Mice , Mice, Inbred C57BL , Mutation , RNA, Messenger/genetics , RNA, Messenger/immunology , Specific Pathogen-Free Organisms , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology , Viral Nonstructural Proteins/genetics , Viral Nonstructural Proteins/immunology , Viral Vaccines/immunology , Zika Virus Infection/virology
4.
Cell ; 165(5): 1081-1091, 2016 May 19.
Article in English | MEDLINE | ID: mdl-27180225

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Fetal Diseases/virology , Placenta Diseases/virology , Pregnancy Complications, Infectious/virology , Zika Virus Infection/pathology , Zika Virus/physiology , Animals , Apoptosis , Brain/embryology , Brain/pathology , Brain/virology , Female , Fetal Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Placenta Diseases/pathology , Pregnancy , Pregnancy Complications, Infectious/pathology , RNA, Viral/isolation & purification , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Zika Virus Infection/virology
5.
Nat Immunol ; 18(11): 1261-1269, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28945244

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Dengue Virus/immunology , Epitopes/immunology , Viral Envelope Proteins/immunology , Zika Virus Infection/immunology , Animals , Brain/immunology , Brain/virology , Chlorocebus aethiops , Cross Reactions/immunology , Dengue Virus/classification , Dengue Virus/metabolism , Female , Fetus/immunology , Fetus/virology , Host-Pathogen Interactions/immunology , Humans , Male , Mice , Neutralization Tests , Pregnancy , Protein Multimerization/immunology , Testis/immunology , Testis/virology , Vero Cells , Viral Envelope Proteins/chemistry , Viral Load/immunology , Zika Virus/immunology , Zika Virus/physiology , Zika Virus Infection/virology
6.
Proc Natl Acad Sci U S A ; 121(28): e2404062121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968109

ABSTRACT

Nutrient sensing and adaptation in the placenta are essential for pregnancy viability and proper fetal growth. Our recent study demonstrated that the placenta adapts to nutrient insufficiency through mechanistic target of rapamycin (mTOR) inhibition-mediated trophoblast differentiation toward syncytiotrophoblasts (STBs), a highly specialized multinucleated trophoblast subtype mediating extensive maternal-fetal interactions. However, the underlying mechanism remains elusive. Here, we unravel the indispensable role of the mTORC1 downstream transcriptional factor TFEB in STB formation both in vitro and in vivo. TFEB deficiency significantly impaired STB differentiation in human trophoblasts and placenta organoids. Consistently, systemic or trophoblast-specific deletion of Tfeb compromised STB formation and placental vascular construction, leading to severe embryonic lethality. Mechanistically, TFEB conferred direct transcriptional activation of the fusogen ERVFRD-1 in human trophoblasts and thereby promoted STB formation, independent of its canonical function as a master regulator of the autophagy-lysosomal pathway. Moreover, we demonstrated that TFEB directed the trophoblast syncytialization response driven by mTOR complex 1 (mTORC1) signaling. TFEB expression positively correlated with the reinforced trophoblast syncytialization in human fetal growth-restricted placentas exhibiting suppressed mTORC1 activity. Our findings substantiate that the TFEB-fusogen axis ensures proper STB formation during placenta development and under nutrient stress, shedding light on TFEB as a mechanistic link between nutrient-sensing machinery and trophoblast differentiation.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Cell Differentiation , Mechanistic Target of Rapamycin Complex 1 , Trophoblasts , Trophoblasts/metabolism , Humans , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Female , Pregnancy , Mice , Animals , Mechanistic Target of Rapamycin Complex 1/metabolism , Placenta/metabolism , Signal Transduction , Autophagy/physiology
7.
Dev Biol ; 493: 29-39, 2023 01.
Article in English | MEDLINE | ID: mdl-36368522

ABSTRACT

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.


Subject(s)
Urinary Tract Infections , Urinary Tract , Female , Mice , Animals , Urinary Bladder , Aging , Homeostasis , Immunity, Innate
8.
J Urol ; 209(5): 928-936, 2023 05.
Article in English | MEDLINE | ID: mdl-36715657

ABSTRACT

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.


Subject(s)
Cystitis , Urinary Tract Infections , Female , Humans , Cystitis/complications , Cystitis/drug therapy , Cystitis/pathology , Lymphoid Tissue/pathology , Pain/pathology , Postmenopause , Retrospective Studies , Urinary Bladder/pathology , Urinary Tract Infections/etiology , Urinary Tract Infections/complications
9.
Nature ; 540(7633): 443-447, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27819683

ABSTRACT

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.


Subject(s)
Antibodies, Neutralizing/immunology , Fetal Diseases/prevention & control , Infectious Disease Transmission, Vertical/prevention & control , Virus Replication/immunology , Zika Virus Infection/immunology , Zika Virus Infection/virology , Zika Virus/growth & development , Zika Virus/immunology , Africa , Americas , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/therapeutic use , Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Antibodies, Viral/therapeutic use , Antibody Specificity , Asia , B-Lymphocytes/immunology , Disease Models, Animal , Epitope Mapping , Female , Fetal Diseases/immunology , Fetal Diseases/virology , Fetus/immunology , Fetus/virology , Humans , Male , Mice , Models, Molecular , Placenta/immunology , Placenta/virology , Pregnancy , Protein Multimerization , Survival Rate , Viral Proteins/chemistry , Viral Proteins/immunology , Viral Vaccines/chemistry , Viral Vaccines/immunology , Zika Virus Infection/pathology
10.
Fetal Pediatr Pathol ; 41(3): 403-412, 2022 Jun.
Article in English | MEDLINE | ID: mdl-33040615

ABSTRACT

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.


Subject(s)
COVID-19 , Pregnancy Complications, Infectious , Case-Control Studies , Female , Humans , Placenta/pathology , Pregnancy , Pregnancy Complications, Infectious/pathology , Pregnancy Trimester, Third , Retrospective Studies , SARS-CoV-2
11.
Am J Obstet Gynecol ; 224(5): 530.e1-530.e17, 2021 05.
Article in English | MEDLINE | ID: mdl-33248136

ABSTRACT

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.


Subject(s)
Coinfection/complications , Gardnerella vaginalis , Placenta Diseases/microbiology , Streptococcal Infections/microbiology , Streptococcus agalactiae , Uterine Diseases/microbiology , Vaginosis, Bacterial/microbiology , Animals , Cytokines/metabolism , Dysbiosis/microbiology , Female , Mice , Microbial Interactions , Microbiota , Placenta/microbiology , Placenta Diseases/metabolism , Placenta Diseases/pathology , Pregnancy , Vagina/microbiology
12.
J Biol Chem ; 293(16): 6022-6038, 2018 04 20.
Article in English | MEDLINE | ID: mdl-29496999

ABSTRACT

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.


Subject(s)
Gene Deletion , Animals , Autoantigens/analysis , Bacterial Infections/genetics , Bacterial Infections/immunology , CRISPR-Cas Systems , Female , Immunity, Innate , Inflammation/genetics , Inflammation/immunology , Male , Membrane Proteins/analysis , Mice , Mycoses/genetics , Mycoses/immunology , Phylogeny , Virus Diseases/genetics , Virus Diseases/immunology
13.
Am J Physiol Renal Physiol ; 316(5): F814-F822, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30724105

ABSTRACT

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.


Subject(s)
Escherichia coli Infections/diet therapy , Iron, Dietary/metabolism , Urinary Bladder/microbiology , Urinary Tract Infections/diet therapy , Uropathogenic Escherichia coli/pathogenicity , Animals , Bacterial Load , Disease Models, Animal , Escherichia coli Infections/metabolism , Escherichia coli Infections/microbiology , Ferritins/metabolism , Hepcidins/genetics , Hepcidins/metabolism , Host-Pathogen Interactions , Interleukin-6/metabolism , Mice, Inbred C57BL , Mice, Knockout , Urinary Bladder/metabolism , Urinary Tract Infections/metabolism , Urinary Tract Infections/microbiology
14.
Hum Reprod ; 34(6): 1106-1116, 2019 06 04.
Article in English | MEDLINE | ID: mdl-31037294

ABSTRACT

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.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Endometriosis/drug therapy , Gastrointestinal Microbiome/drug effects , Metronidazole/administration & dosage , Peritoneal Diseases/drug therapy , Animals , Disease Models, Animal , Disease Progression , Endometriosis/microbiology , Endometriosis/pathology , Endometrium/pathology , Fecal Microbiota Transplantation/adverse effects , Feces/microbiology , Female , Gastrointestinal Microbiome/physiology , Humans , Mice , Peritoneal Diseases/microbiology , Peritoneal Diseases/pathology
15.
N Engl J Med ; 383(21): 2080-2082, 2020 11 19.
Article in English | MEDLINE | ID: mdl-33207100
16.
J Urol ; 209(5): 949, 2023 05.
Article in English | MEDLINE | ID: mdl-36815426
17.
Dev Dyn ; 246(4): 336-343, 2017 04.
Article in English | MEDLINE | ID: mdl-28109014

ABSTRACT

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.


Subject(s)
Regeneration , Urologic Neoplasms/pathology , Urothelium/growth & development , Animals , Humans , Signal Transduction , Urinary Bladder/physiology , Urologic Diseases/physiopathology , Urothelium/injuries , Urothelium/physiology
18.
Nature ; 472(7341): 110-4, 2011 Apr 07.
Article in English | MEDLINE | ID: mdl-21389986

ABSTRACT

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.


Subject(s)
Epithelial Cells/cytology , Hedgehog Proteins/metabolism , Regeneration/physiology , Stem Cells/cytology , Urinary Bladder/cytology , Wnt Proteins/metabolism , Animals , Cell Lineage , Cell Proliferation , Epithelial Cells/metabolism , Feedback, Physiological , Female , Fibroblast Growth Factors/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Mice , Organoids/cytology , Signal Transduction , Stem Cells/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Urinary Bladder/drug effects , Urinary Bladder/injuries , Urinary Bladder/metabolism , Urinary Bladder Diseases/chemically induced , Urinary Bladder Diseases/metabolism , Urinary Bladder Diseases/microbiology , Urinary Bladder Diseases/pathology , Uropathogenic Escherichia coli/physiology , Urothelium/cytology , Zinc Finger Protein GLI1
20.
Mo Med ; 114(3): 168-170, 2017.
Article in English | MEDLINE | ID: mdl-30228574

ABSTRACT

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.


Subject(s)
Abortion, Spontaneous/virology , Fetal Growth Retardation/virology , Microcephaly/virology , Placenta/virology , Zika Virus Infection/transmission , Abortion, Spontaneous/epidemiology , Animals , Female , Fetal Death/etiology , Fetal Growth Retardation/epidemiology , Fetus , Humans , Infectious Disease Transmission, Vertical/prevention & control , Mice , Microcephaly/epidemiology , Models, Animal , Pregnancy , Pregnancy Complications, Infectious/epidemiology , Trophoblasts/virology , World Health Organization/organization & administration , Zika Virus/genetics , Zika Virus/isolation & purification , Zika Virus Infection/complications
SELECTION OF CITATIONS
SEARCH DETAIL