Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Pathog ; 20(9): e1012578, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39321205

ABSTRACT

The MtrCDE efflux pump of Neisseria gonorrhoeae exports a wide range of antimicrobial compounds that the gonococcus encounters at mucosal surfaces during colonization and infection and is a known gonococcal virulence factor. Here, we evaluate the role of this efflux pump system in strain FA1090 during in vivo human male urethral infection with N. gonorrhoeae using a controlled human infection model. With the strategy of competitive infections initiated with mixtures of wild-type FA1090 and an isogenic mutant FA1090 strain that does not contain a functional MtrCDE pump, we found that the presence of the efflux pump is not required for an infection to be established in the human male urethra. This finding contrasts with previous studies of in vivo infection in the lower genital tract of female mice, which demonstrated that mutant gonococci of a different strain (FA19) lacking a functional MtrCDE pump had a significantly reduced fitness compared to their wild-type parental FA19 strain. To determine if these conflicting results are due to strain or human vs. mouse differences, we conducted a series of systematic competitive infections in female mice with the same FA1090 strains as in humans, and with FA19 strains, including mutants that do not assemble a functional MtrCDE efflux pump. Our results indicate the fitness advantage provided by the MtrCDE efflux pump during infection of mice is strain dependent. Owing to the equal fitness of the two FA1090 strains in men, our experiments also demonstrated the presence of a colonization bottleneck of N. gonorrhoeae in the human male urethra, which may open a new area of inquiry into N. gonorrhoeae infection dynamics and control. TRIAL REGISTRATION. Clinicaltrials.gov NCT03840811.


Subject(s)
Gonorrhea , Neisseria gonorrhoeae , Animals , Female , Humans , Male , Mice , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Gonorrhea/microbiology , Membrane Transport Proteins/metabolism , Membrane Transport Proteins/genetics , Neisseria gonorrhoeae/genetics , Neisseria gonorrhoeae/pathogenicity , Neisseria gonorrhoeae/metabolism , Reproductive Tract Infections/microbiology , Reproductive Tract Infections/metabolism , Urethra/microbiology
2.
J Immunol ; 205(11): 3037-3049, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33087404

ABSTRACT

Chlamydia trachomatis infection of the female genital tract can lead to irreversible fallopian tube scarring. In the mouse model of genital infection using Chlamydia muridarum, IL-1R signaling plays a critical role in oviduct tissue damage. In this study, we investigated the pathologic role of IL-1α, one of the two proinflammatory cytokines that bind to IL-1R. Il1a-/- mice infected with C. muridarum cleared infection at their cervix at the same rate as wild-type (WT) mice, but were significantly protected from end point oviduct damage and fibrosis. The contribution of IL-1α to oviduct pathology was more dramatic than observed in mice deficient for IL-1ß. Although chlamydial burden was similar in WT and Il1a-/- oviduct during peak days of infection, levels of IL-1ß, IL-6, CSF3, and CXCL2 were reduced in Il1a-/- oviduct lysates. During infection, Il1a-/- oviducts and uterine horns exhibited reduced neutrophil infiltration, and this reduction persisted after the infection resolved. The absence of IL-1α did not compromise CD4 T cell recruitment or function during primary or secondary chlamydial infection. IL-1α is expressed predominantly by luminal cells of the genital tract in response to infection, and low levels of expression persisted after the infection cleared. Ab-mediated depletion of IL-1α in WT mice prevented infection-induced oviduct damage, further supporting a key role for IL-1α in oviduct pathology.


Subject(s)
Chlamydia Infections/metabolism , Genitalia, Female/metabolism , Interleukin-1alpha/metabolism , Oviducts/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , Cervix Uteri/metabolism , Cervix Uteri/microbiology , Chlamydia Infections/microbiology , Chlamydia muridarum/pathogenicity , Disease Models, Animal , Female , Genitalia, Female/microbiology , Interleukin-1beta/metabolism , Mice , Mice, Inbred C57BL , Neutrophil Infiltration/physiology , Oviducts/microbiology , Reproductive Tract Infections/metabolism , Reproductive Tract Infections/microbiology
3.
Int J Mol Sci ; 23(16)2022 Aug 21.
Article in English | MEDLINE | ID: mdl-36012708

ABSTRACT

In mammals, ß-defensins have been reported to play pivotal roles in sperm protection and fertilization. However, the function and mechanism of porcine ß-defensin 129 (pBD129) in the sperm remain unclear. Here, we demonstrate that pBD129 is a glycosylated protein and broadly exists in accessory sex glands and coats the sperm surface. We inhibited the pBD129 protein on the sperm surface with an anti-pBD129 antibody and found that sperm motility was not significantly affected; however, sperm acrosome integrity and tyrosine phosphorylation levels increased significantly with time (p < 0.05) during capacitation. These changes were accompanied by an increase in sperm Ca2+ influx, resulting in a significantly reduced in vitro fertilization cleavage rate (p < 0.05). Further investigation revealed that treatment with recombinant pBD129 markedly restored the sperm motility in semen contaminated with Escherichia coli. The results suggest that pBD129 is not only associated with poor sperm motility after genital tract infection but can also protect the spermatozoa from premature capacitation, which may be beneficial for semen preservation.


Subject(s)
Reproductive Tract Infections , beta-Defensins , Acrosome Reaction , Animals , Male , Mammals , Reproductive Tract Infections/metabolism , Semen , Sperm Capacitation , Spermatozoa/metabolism , Swine , beta-Defensins/metabolism
4.
Infect Immun ; 89(10): e0007221, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34125599

ABSTRACT

Genital infections with Chlamydia trachomatis can lead to uterine and oviduct tissue damage in the female reproductive tract. Neutrophils are strongly associated with tissue damage during chlamydial infection, while an adaptive CD4 T cell response is necessary to combat infection. Activation of triggering receptor expressed on myeloid cells-1 (TREM-1) on neutrophils has previously been shown to induce and/or enhance degranulation synergistically with Toll-like receptor (TLR) signaling. Additionally, TREM-1 can promote neutrophil transepithelial migration. In this study, we sought to determine the contribution of TREM-1,3 to immunopathology in the female mouse genital tract during Chlamydia muridarum infection. Relative to control mice, trem1,3-/- mice had no difference in chlamydial burden or duration of lower-genital-tract infection. We also observed a similar incidence of hydrosalpinx 45 days postinfection in trem1,3-/- compared to wild-type (WT) mice. However, compared to WT mice, trem1,3-/- mice developed significantly fewer hydrometra in uterine horns. Early in infection, trem1,3-/- mice displayed a notable decrease in the number of uterine glands containing polymorphonuclear cells and uterine horn lumens had fewer neutrophils, with increased granulocyte colony-stimulating factor (G-CSF). trem1,3-/- mice also had reduced erosion of the luminal epithelium. These data indicate that TREM-1,3 contributes to transepithelial neutrophil migration in the uterus and uterine glands, promoting the occurrence of hydrometra in infected mice.


Subject(s)
Chlamydia Infections/immunology , Chlamydia muridarum/immunology , Receptors, Immunologic/immunology , Triggering Receptor Expressed on Myeloid Cells-1/immunology , Uterus/immunology , Adaptive Immunity/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/microbiology , Cell Movement/immunology , Chlamydia Infections/metabolism , Chlamydia Infections/microbiology , Chlamydia trachomatis/immunology , Disease Models, Animal , Epithelium/immunology , Epithelium/metabolism , Epithelium/microbiology , Female , Genitalia, Female/immunology , Genitalia, Female/metabolism , Genitalia, Female/microbiology , Mice , Mice, Inbred C57BL , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/microbiology , Oviducts/immunology , Oviducts/metabolism , Oviducts/microbiology , Receptors, Immunologic/metabolism , Reproductive Tract Infections/immunology , Reproductive Tract Infections/metabolism , Reproductive Tract Infections/microbiology , Triggering Receptor Expressed on Myeloid Cells-1/metabolism , Uterus/metabolism , Uterus/microbiology
5.
Reprod Biol Endocrinol ; 19(1): 97, 2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34183027

ABSTRACT

Recent years have seen a rising incidence of male infertility, mostly caused by the decline of sperm quality. The ratio of infertile males to infertile females has escalated from 3:7 in 2013 to current 5:5, which turns male infertility into the research focus of reproductive medicine. This study aimed to clarify the effect of reproductive tract infection by ureaplasma urealyticum (UU) and chlamydia trachomatis (CT) on the DNA integrity and routine semen parameters of infertile males. A retrospective study was performed. A total of 259 infertile males who were treated at the Andrological Laboratory Examination and Reproductive Medicine Center in our hospital were analyzed. qRT-PCR was used to examine the infection status of CT and UU. According to the eligibility criteria, we evaluated the semen parameters and biochemical data of 253 men. Based on the results of PCR, the subjects were divided into four groups: Group I (CT positive, 63 cases), Group II (UU positive, 60 cases), Group III (CT positive and UU positive, 62 cases), and Group IV (no infection, 68 cases). DNA fragmentation index (DFI), sperm count, vitality and morphology, elastase level, seminal plasma malondialdehyde (MDA), and total antioxidant capacity (TAC) were assessed. Compared to Group IV, three groups (Group I, Group II and Group III) showed difference in semen volume, proportion of sperm with normal morphology, sperm motility, progressive motility, and vitality (P < 0.05). Compared to Group IV, Group II and Group III showed difference in DFI (P < 0.05). Compared to Group IV, Group II and Group III showed difference in elastase level (P < 0.05). VCL, VSL, VAP, WOB, ROS, TM, HDS showed differences between groups of abnormal/normal WBC (*P < 0.01).UU infection significantly increased the level of seminal leukocytes only in Group II, but not in the other three groups, indicating that UU is a factor to increase the level of seminal leukocytes. Compared with the normal leukocyte group, there were significant differences in total motility, forward motility and normal sperm ratio between the two groups. The proportion of sperm with abnormal morphology (mostly in the head) showed obvious difference between groups of high and normal seminal leukocytic levels. At the same time, in this study, SCGE and SCD verified that leukocytes could damage sperm DNA by increasing ROS, which ultimately affects male fertility.


Subject(s)
DNA Fragmentation , Infertility, Male/metabolism , Oxidative Stress/physiology , Reproductive Tract Infections/metabolism , Semen Analysis/methods , Semen/metabolism , Adolescent , Adult , Humans , Infertility, Male/genetics , Male , Reproductive Tract Infections/genetics , Sperm Motility/physiology , Young Adult
6.
Am J Physiol Endocrinol Metab ; 318(6): E981-E994, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32315215

ABSTRACT

Chlamydia trachomatis infection is a primary cause of reproductive tract diseases including infertility. Previous studies showed that this infection alters physiological activities in mouse oviducts. Whether this occurs in the uterus and cervix has never been investigated. This study characterized the physiological activities of the uterine horn and the cervix in a Chlamydia muridarum (Cmu)-infected mouse model at three infection time points of 7, 14, and 21 days postinfection (dpi). Cmu infection significantly decreased contractile force of spontaneous contraction in the cervix (7 and 14 dpi; P < 0.001 and P < 0.05, respectively), but this effect was not observed in the uterine horn. The responses of the uterine horn and cervix to oxytocin were significantly altered by Cmu infection at 7 dpi (P < 0.0001), but such responses were attenuated at 14 and 21 dpi. Cmu infection increased contractile force to prostaglandin (PGF2α) by 53-83% in the uterine horn. This corresponded with the increased messenger ribonucleic acid (mRNA) expression of Ptgfr that encodes for its receptor. However, Cmu infection did not affect contractions of the uterine horn and cervix to PGE2 and histamine. The mRNA expression of Otr and Ptger4 was inversely correlated with the mRNA expression of Il1b, Il6 in the uterine horn of Cmu-inoculated mice (P < 0.01 to P < 0.001), suggesting that the changes in the Otr and Ptger4 mRNA expression might be linked to the changes in inflammatory cytokines. Lastly, this study also showed a novel physiological finding of the differential response to PGE2 in mouse uterine horn and cervix.


Subject(s)
Chlamydia Infections/physiopathology , Chlamydia muridarum , Myometrium/physiopathology , Reproductive Tract Infections/physiopathology , Uterine Contraction/physiology , Uterus/physiopathology , Animals , Cervix Uteri/metabolism , Cervix Uteri/physiopathology , Chlamydia Infections/genetics , Chlamydia Infections/immunology , Chlamydia Infections/metabolism , Cytokines/genetics , Dinoprost/pharmacology , Dinoprostone/pharmacology , Female , Gene Expression Regulation , Histamine/pharmacology , Histamine Agonists/pharmacology , Interleukin-1beta/genetics , Interleukin-6/genetics , Mice , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/drug effects , Muscle, Smooth/physiopathology , Myometrium/drug effects , Myometrium/metabolism , Oviducts/pathology , Oxytocics/pharmacology , RNA, Messenger/metabolism , Receptors, Oxytocin/genetics , Receptors, Prostaglandin/genetics , Receptors, Prostaglandin E, EP4 Subtype/genetics , Reproductive Tract Infections/genetics , Reproductive Tract Infections/immunology , Reproductive Tract Infections/metabolism , Uterine Contraction/drug effects , Uterus/metabolism
7.
Gynecol Endocrinol ; 35(9): 796-802, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30935248

ABSTRACT

Why most women can clear human papillomavirus (HPV) infections while others can develop permanent infections. The stimulation of immunotolerance of the immune system of the host by the persistent HPV infection may be the answer to this question. Interleukin-33 (IL-33) may play a role in the pathogenesis of HPV infection, this hypothesis was thought to be due to the rapid release of IL-33 from damaged cells following tissue damage, necrosis, and activation of the inflammasome. Thus, in this study, the role of IL-33/suppressor of tumorigenicity 2 (ST2) was emphasized in HPV positive and HPV negative cervical tissues. A total of 80 were assessed. The reduced levels of IL-33 and ST2 are associated with cervical HPV infections. There was a statistically significant 42% positive correlation between IL-33 and ST2 in the HPV-positive group. Surprisingly, our data showed no significant difference between the expression levels of IL-33 or ST2 and working status, type of delivery, pre- and post-operative pathology, cigarette, educational status, locality, birth control method, gynecological, and colposcopic findings. We found that as a result of our study; low IL-33 and ST2 levels were associated with HPV infections.


Subject(s)
Interleukin-1 Receptor-Like 1 Protein/physiology , Interleukin-33/physiology , Papillomavirus Infections , Reproductive Tract Infections , Adult , Aged , Biopsy , Cervix Uteri/metabolism , Cervix Uteri/pathology , Cervix Uteri/virology , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , Humans , Interleukin-1 Receptor-Like 1 Protein/blood , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/blood , Interleukin-33/metabolism , Middle Aged , Papillomaviridae/genetics , Papillomaviridae/isolation & purification , Papillomavirus Infections/epidemiology , Papillomavirus Infections/metabolism , Papillomavirus Infections/pathology , Reproductive Tract Infections/epidemiology , Reproductive Tract Infections/metabolism , Reproductive Tract Infections/virology , Uterine Cervical Neoplasms/epidemiology , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virology , Vaginal Smears , Young Adult , Uterine Cervical Dysplasia/epidemiology , Uterine Cervical Dysplasia/metabolism , Uterine Cervical Dysplasia/pathology , Uterine Cervical Dysplasia/virology
8.
Mol Pharm ; 15(4): 1534-1547, 2018 04 02.
Article in English | MEDLINE | ID: mdl-29481088

ABSTRACT

The need for more versatile technologies to deliver antiviral agents to the female reproductive tract (FRT) has spurred the development of on-demand and sustained-release platforms. Electrospun fibers (EFs), in particular, have recently been applied to FRT delivery, resulting in an alternative dosage form with the potential to provide protection and therapeutic effect against a variety of infection types. However, a multitude of fabrication parameters, as well as the resulting complexities of solvent-drug, drug-polymer, and solvent-polymer interactions, are known to significantly impact the loading and release of incorporated agents. Numerous processing parameters, in addition to their combined interactions, can hinder the iterative development of fiber formulations to achieve optimal release for particular durations, doses, and polymer-drug types. The experimental effort to design and develop EFs could benefit from mathematical analysis and computational simulation that predictively evaluate combinations of parameters to meet product design needs. Here, existing modeling efforts are leveraged to develop a simulation platform that correlates and predicts the delivery of relevant small molecule antivirals from EFs that have been recently applied to target sexually transmitted infections (STIs). A pair of mathematical models is coupled to simulate the release of two structurally similar small molecule antiretroviral reverse transcriptase inhibitors, Tenofovir (TFV) and Tenofovir disoproxil fumarate (TDF), from poly(lactic- co-glycolic acid) (PLGA) EFs, and to evaluate how changes in the system parameters affect the distribution of encapsulated agent in a three-compartment model of the vaginal epithelium. The results indicate that factors such as fiber diameter, mesh thickness, antiviral diffusivity, and fiber geometry can be simulated to create an accurate model that distinguishes the different release patterns of TFV and TDF from EFs, and that enables detailed evaluation of the associated pharmacokinetics. This simulation platform offers a basis with which to further study EF parameters and their effect on antiviral release and pharmacokinetics in the FRT.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Genitalia, Female/metabolism , Polyesters/metabolism , Reproductive Tract Infections/metabolism , Anti-HIV Agents/pharmacology , Computer Simulation , Diffusion , Epithelium/metabolism , Epithelium/virology , Female , Genitalia, Female/virology , HIV Infections/drug therapy , HIV Infections/metabolism , HIV-1/drug effects , Humans , Reproductive Tract Infections/drug therapy , Reproductive Tract Infections/virology , Reverse Transcriptase Inhibitors/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacology , Tenofovir/pharmacokinetics , Tenofovir/pharmacology
9.
J Virol ; 90(9): 4258-4261, 2016 May.
Article in English | MEDLINE | ID: mdl-26865718

ABSTRACT

Respiratory virus infections are common but generally self-limiting infections in healthy individuals. Although early clinical studies reported low detection rates, the development of molecular diagnostic techniques by PCR has led to an increased recognition that respiratory virus infections are associated with morbidity and acute exacerbations of chronic lung diseases, such as cystic fibrosis (CF). The airway epithelium is the first barrier encountered by respiratory viruses following inhalation and the primary site of respiratory viral replication. Here, we describe how the airway epithelial response to respiratory viral infections contributes to disease progression in patients with CF and other chronic lung diseases, including the role respiratory viral infections play in bacterial acquisition in the CF patient lung.


Subject(s)
Cystic Fibrosis/complications , Opportunistic Infections/etiology , Reproductive Tract Infections/etiology , Respiratory Mucosa/virology , Virus Diseases/etiology , Animals , Bacterial Infections/etiology , Bacterial Infections/metabolism , Bacterial Infections/pathology , Chronic Disease , Coinfection , Disease Progression , Humans , Lung Diseases/complications , Opportunistic Infections/metabolism , Opportunistic Infections/pathology , Reproductive Tract Infections/metabolism , Reproductive Tract Infections/pathology , Virus Diseases/metabolism , Virus Diseases/pathology
10.
Infect Immun ; 84(8): 2324-2335, 2016 08.
Article in English | MEDLINE | ID: mdl-27271740

ABSTRACT

Pseudomonas aeruginosa is a leading cause of hospital-acquired pneumonia and chronic lung infections in cystic fibrosis patients. Iron is essential for bacterial growth, and P. aeruginosa expresses multiple iron uptake systems, whose role in lung infection deserves further investigation. P. aeruginosa Fe(3+) uptake systems include the pyoverdine and pyochelin siderophores and two systems for heme uptake, all of which are dependent on the TonB energy transducer. P. aeruginosa also has the FeoB transporter for Fe(2+) acquisition. To assess the roles of individual iron uptake systems in P. aeruginosa lung infection, single and double deletion mutants were generated in P. aeruginosa PAO1 and characterized in vitro, using iron-poor media and human serum, and in vivo, using a mouse model of lung infection. The iron uptake-null mutant (tonB1 feoB) and the Fe(3+) transport mutant (tonB1) did not grow aerobically under low-iron conditions and were avirulent in the mouse model. Conversely, the wild type and the feoB, hasR phuR (heme uptake), and pchD (pyochelin) mutants grew in vitro and caused 60 to 90% mortality in mice. The pyoverdine mutant (pvdA) and the siderophore-null mutant (pvdA pchD) grew aerobically in iron-poor media but not in human serum, and they caused low mortality in mice (10 to 20%). To differentiate the roles of pyoverdine in iron uptake and virulence regulation, a pvdA fpvR double mutant defective in pyoverdine production but expressing wild-type levels of pyoverdine-regulated virulence factors was generated. Deletion of fpvR in the pvdA background partially restored the lethal phenotype, indicating that pyoverdine contributes to the pathogenesis of P. aeruginosa lung infection by combining iron transport and virulence-inducing capabilities.


Subject(s)
Iron/metabolism , Pseudomonas Infections/metabolism , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/physiology , Reproductive Tract Infections/metabolism , Reproductive Tract Infections/microbiology , Animals , Disease Models, Animal , Humans , Mice , Mutation , Oligopeptides/metabolism , Phenotype , Pseudomonas aeruginosa/genetics , Signal Transduction , Transferrin/metabolism , Virulence
11.
Virol J ; 13: 24, 2016 Feb 06.
Article in English | MEDLINE | ID: mdl-26852031

ABSTRACT

Human coronaviruses (HCoVs) are large RNA viruses that infect the human respiratory tract. The emergence of both Severe Acute Respiratory Syndrome and Middle East Respiratory syndrome CoVs as well as the yearly circulation of four common CoVs highlights the importance of elucidating the different mechanisms employed by these viruses to evade the host immune response, determine their tropism and identify antiviral compounds. Various animal models have been established to investigate HCoV infection, including mice and non-human primates. To establish a link between the research conducted in animal models and humans, an organotypic human airway culture system, that recapitulates the human airway epithelium, has been developed. Currently, different cell culture systems are available to recapitulate the human airways, including the Air-Liquid Interface (ALI) human airway epithelium (HAE) model. Tracheobronchial HAE cultures recapitulate the primary entry point of human respiratory viruses while the alveolar model allows for elucidation of mechanisms involved in viral infection and pathogenesis in the alveoli. These organotypic human airway cultures represent a universal platform to study respiratory virus-host interaction by offering more detailed insights compared to cell lines. Additionally, the epidemic potential of this virus family highlights the need for both vaccines and antivirals. No commercial vaccine is available but various effective antivirals have been identified, some with potential for human treatment. These morphological airway cultures are also well suited for the identification of antivirals, evaluation of compound toxicity and viral inhibition.


Subject(s)
Coronavirus Infections/virology , Coronavirus/physiology , Host-Pathogen Interactions , Reproductive Tract Infections/virology , Animals , Coronavirus/classification , Coronavirus Infections/immunology , Coronavirus Infections/metabolism , Coronavirus Infections/therapy , Disease Models, Animal , Humans , Immunity, Innate , Receptors, Virus/chemistry , Receptors, Virus/metabolism , Reproductive Tract Infections/immunology , Reproductive Tract Infections/metabolism , Reproductive Tract Infections/therapy , Respiratory Mucosa/immunology , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Viral Tropism
12.
Immunology ; 146(4): 557-67, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26302175

ABSTRACT

T helper type 17 (Th17) cells play an important role in immunity to fungal and bacterial pathogens, although their role in the female genital tract, where exposure to these pathogens is common, is not well understood. We investigated the relationship between female genital tract infections, cervicovaginal interleukin-17 (IL-17) concentrations and Th17 cell frequencies. Forty-two cytokines were measured in cervicovaginal lavages from HIV-uninfected and HIV-infected women. Frequencies of Th17 cells (CD3(+) CD4(+) IL-17a(+)) were evaluated in cervical cytobrushes and blood by flow cytometry. Women were screened for Chlamydia trachomatis, Neisseria gonorrhoeae, Mycoplasma genitalium, Trichomonas vaginalis and herpes simplex virus 2 by PCR, and candidal infections and bacterial vaginosis by Gram stain. Women with bacterial sexually transmitted infections (STIs), specifically chlamydia and gonorrhoea, had higher genital IL-17 concentrations than women with no STI, whereas women with candidal pseudohyphae/spores had lower IL-17 concentrations compared with women without candidal infections. Viral STIs (herpes simplex virus 2 and HIV) were not associated with significant changes in genital IL-17 concentrations. Genital IL-17 concentrations correlated strongly with other inflammatory cytokines and growth factors. Although Th17 cells were depleted from blood during HIV infection, cervical Th17 cell frequencies were similar in HIV-uninfected and HIV-infected women. Cervical Th17 cell frequencies were also not associated with STIs or candida, although few women had a STI. These findings suggest that IL-17 production in the female genital tract is induced in response to bacterial but not viral STIs. Decreased IL-17 associated with candidal infections suggests that candida may actively suppress IL-17 production or women with dampened IL-17 responses may be more susceptible to candidal outgrowth.


Subject(s)
Interleukin-17/biosynthesis , Mucous Membrane/immunology , Mucous Membrane/metabolism , Reproductive Tract Infections/immunology , Reproductive Tract Infections/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Adult , Bacterial Infections/immunology , Bacterial Infections/metabolism , Bacterial Infections/microbiology , CD4 Lymphocyte Count , Candidiasis/immunology , Candidiasis/metabolism , Candidiasis/microbiology , Cervix Uteri , Cytokines/biosynthesis , Female , Humans , Interleukin-17/blood , Mucous Membrane/microbiology , Reproductive Tract Infections/microbiology , Sexually Transmitted Diseases/immunology , Sexually Transmitted Diseases/metabolism , Sexually Transmitted Diseases/microbiology , Sexually Transmitted Diseases/virology , Vagina , Young Adult
13.
Tissue Antigens ; 83(4): 273-85, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24641504

ABSTRACT

S5D-SRCRB is a novel mouse secretory glycoprotein belonging to the ancient and highly conserved scavenger receptor cysteine-rich superfamily of protein receptors. Available evidence indicates that S5D-SRCRB interacts with conserved microbial cell wall components, as well as with some endogenous proteins, and presents a restricted tissue expression pattern. This study further analyzes the expression of S5D-SRCRB along the mouse urogenital tract. Immunohistochemical staining for S5D-SRCRB was observed in spermatocytes from seminiferous tubules and in the epithelial surface from urethra and bladder, as well as in kidney tubules, mainly from medulla and papilla. Double stainings showed that S5D-SRCRB is expressed in both principal (P) and intercalated (IC) cells from renal collecting ducts (CD). By using an in vitro cell model of IC cell differentiation, preferential expression of S5D-SRCRB was observed in the apical border of terminally differentiated IC. Colocalization of S5D-SRCRB with galectin-3 (Gal-3) was also observed in kidney and bladder, but not in testis, supporting concurrent biochemical studies demonstrating the carbohydrate-dependent interaction of Gal-3 and S5D-SRCRB. Furthermore, upregulation of S5D-SRCRB expression was observed in in vitro and in vivo models of bacterial aggression, reinforcing the emerging view that CD, and specially IC, are important players in innate defense of the urinary tract against infection. Taken together, the results indicate that S5D-SRCRB is an integral component of the urogenital tract involved in innate immune functions.


Subject(s)
Gene Expression Regulation/immunology , Immunity, Innate , Scavenger Receptors, Class B/immunology , Urethra/immunology , Urinary Bladder/immunology , Animals , Mice , Mice, Inbred BALB C , Reproductive Tract Infections/immunology , Reproductive Tract Infections/metabolism , Scavenger Receptors, Class B/biosynthesis , Urethra/metabolism , Urinary Bladder/metabolism , Urinary Tract Infections/immunology , Urinary Tract Infections/metabolism
14.
Am J Reprod Immunol ; 89(2): e13588, 2023 02.
Article in English | MEDLINE | ID: mdl-35771685

ABSTRACT

PROBLEM: Surfactant protein D (SP-D), a multimeric collectin expressed by testicular mucosal epithelia and is positively regulated by testosterone. It exerts antimicrobial effects, modulates inflammation and rescued spermatogenesis in a murine model. Various cytokines and chemokines, including MCP-1, play a key role in regulating the inflammation in rat and human testis. The study aimed to investigate the role of SP-D and involvement of chemokines and cytokines in the male infertility associated with urogenital infections or inflammation. METHOD OF STUDY: The cross-sectional study evaluated levels of SP-D, testosterone, estradiol and the cytokines/chemokines including MCP-1 in the serum and semen samples of fertile and infertile Indian men with and without urogenital infections/inflammation (n = 76). RESULTS: Both fertile and infertile males with urogenital infection/inflammation had significantly lower levels of SP-D and higher levels of the chemokine, Monocyte chemoattractant protein 1 (MCP-1) in the serum and seminal plasma. Seminal plasma of these males exhibited significantly higher proportion of proteolytically degraded forms of SP-D. The serum SP-D levels positively correlated with testosterone/estradiol (TE) ratio. There was no significant correlation between the SP-D levels in seminal plasma and sperm count/motility. With a significant area under the Receiver Operating Characteristic curves, the serum and seminal plasma SP-D levels exhibited significant potential to predict infertility with high sensitivity and specificity in men with genital infections/inflammation. CONCLUSIONS: The circulating and seminal plasma SP-D levels were decreased in men with urogenital infection and inflammation. This could be due to their engagement at the site of infection, dysregulated expression owing to the altered hormonal profile and increased proteolytic degradation.


Subject(s)
Infertility, Male , Reproductive Tract Infections , Humans , Male , Animals , Mice , Rats , Semen/metabolism , Pulmonary Surfactant-Associated Protein D , Chemokine CCL2/metabolism , Reproductive Tract Infections/metabolism , Cross-Sectional Studies , Testis/metabolism , Testosterone/metabolism , Inflammation/metabolism , Chemokines/metabolism , Estradiol/metabolism
15.
Carbohydr Polym ; 261: 117919, 2021 Jun 01.
Article in English | MEDLINE | ID: mdl-33766328

ABSTRACT

Vaginal administration is a promising route for the local treatment of infectious vaginal diseases since it can bypass the first-pass metabolism, drug interactions, and adverse effects. However, the commercial products currently available for topical vulvovaginal treatment have low acceptability and do not adequately explore this route. Mucoadhesive systems can optimize the efficacy of drugs administered by this route to increase the retention time of the drug in the vaginal environment. Several polymers are used to develop mucoadhesive systems, among them chitosan, a natural polymer that is highly biocompatible and technologically versatile. Thus, the present review aimed to analyze the studies that used chitosan to develop mucoadhesive systems for the treatment of local vaginal infections. These studies demonstrated that chitosan as a component of mucoadhesive drug delivery systems (DDS) is a promising device for the treatment of vaginal infectious diseases, due to the intrinsic antimicrobial activity of this biopolymer and because it does not interfere with the effectiveness of the drugs used for the treatment.


Subject(s)
Anti-Infective Agents, Local/administration & dosage , Chitosan/chemistry , Drug Carriers , Reproductive Tract Infections/drug therapy , Vaginal Diseases/drug therapy , Administration, Intravaginal , Anti-Infective Agents, Local/pharmacokinetics , Biocompatible Materials/chemical synthesis , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacokinetics , Chitosan/chemical synthesis , Chitosan/pharmacokinetics , Drug Carriers/chemical synthesis , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Delivery Systems , Female , Humans , Materials Testing , Reproductive Tract Infections/metabolism , Vaginal Diseases/metabolism
16.
Diabetes Metab Syndr ; 14(6): 1889-1894, 2020.
Article in English | MEDLINE | ID: mdl-33002780

ABSTRACT

BACKGROUND AND AIMS: The link between diabetes and increased risk of infectious disease has long been recognized, but has re-entered sharp focus following the COVID-19 pandemic. METHODS: A literature search was conducted in PubMed for articles in English on diabetes and infection. RESULTS: Diabetes predisposes to infections through alterations in innate and acquired immune defenses. Outcomes of infection are worse in people with uncontrolled diabetes, and infection can worsen hyperglycemia in hitherto well controlled diabetes (bidirectional relationship). Diabetes does not increase the risk of infection with COVID-19 per se, but predisposes to severe disease and poor outcomes. COVID-19 has also been linked to deterioration of glycemic control as well as new-onset diabetes. CONCLUSIONS: Clinicians caring for people with diabetes should be aware of the increased risk of infections in this population, as well as the possibility of worsening hyperglycemia. A holistic approach with frequent monitoring of blood glucose levels and appropriate titration of medications, along with close attention to nutritional status, is essential to ensure the best possible outcomes.


Subject(s)
COVID-19/epidemiology , Diabetes Mellitus/epidemiology , Tuberculosis, Pulmonary/epidemiology , Adaptive Immunity/immunology , Blood Glucose/metabolism , COVID-19/immunology , COVID-19/metabolism , Diabetes Mellitus/immunology , Diabetes Mellitus/metabolism , Glycemic Control , Humans , Immunity, Innate/immunology , India/epidemiology , Infections/epidemiology , Infections/immunology , Infections/metabolism , Reproductive Tract Infections/epidemiology , Reproductive Tract Infections/immunology , Reproductive Tract Infections/metabolism , Respiratory Tract Infections/epidemiology , Respiratory Tract Infections/immunology , Respiratory Tract Infections/metabolism , Risk Factors , SARS-CoV-2 , Severity of Illness Index , Skin Diseases, Bacterial/epidemiology , Skin Diseases, Bacterial/immunology , Skin Diseases, Bacterial/metabolism , Soft Tissue Infections/epidemiology , Soft Tissue Infections/immunology , Soft Tissue Infections/metabolism , Tuberculosis, Pulmonary/immunology , Tuberculosis, Pulmonary/metabolism , Urinary Tract Infections/epidemiology , Urinary Tract Infections/immunology , Urinary Tract Infections/metabolism
17.
Eur J Clin Nutr ; 73(11): 1464-1472, 2019 11.
Article in English | MEDLINE | ID: mdl-31168085

ABSTRACT

BACKGROUND/OBJECTIVES: The iron-binding affinity of vaginal lactoferrin (Lf) reduces iron available to genital pathogens. We describe host reproductive, nutritional, infection and iron biomarker profiles affecting vaginal Lf concentration in young nulliparous and primigravid women in Burkina Faso. SUBJECTS/METHODS: Vaginal eluates from women who had participated in a randomized, controlled periconceptional iron supplementation trial were used to measure Lf using a competitive double-sandwich ELISA. For this analysis samples from both trial arms were combined and pregnant and non-pregnant cohorts compared. Following randomization Lf was measured after 18 months (end assessment) for women remaining non-pregnant, and at two antenatal visits for those becoming pregnant. Associations between log Lf levels and demographic, anthropometric, infection and iron biomarker variables were assessed using linear mixed models. RESULTS: Lf samples were available for 712 non-pregnant women at end assessment and for 303 women seen at an antenatal visit. Lf concentrations of pregnant women were comparable to those of non-pregnant, sexually active women. Lf concentration increased with mid-upper-arm circumference, (P = 0.047), body mass index (P = 0.018), Trichomonas vaginalis (P < 0.001) infection, bacterial vaginosis (P < 0.001), serum C-reactive protein (P = 0.048) and microbiota community state types III/IV. Adjusted Lf concentration was positively associated with serum hepcidin (P = 0.047), serum ferritin (P = 0.018) and total body iron stores (P = 0.042). There was evidence that some women maintained persistently high or low Lf concentrations from before, and through, pregnancy. CONCLUSION: Lf concentrations increased with genital infection, higher BMI, MUAC, body iron stores and hepcidin, suggesting nutritional and iron status influence homeostatic mechanisms controlling vaginal Lf responses.


Subject(s)
Iron/blood , Lactoferrin/analysis , Reproductive Tract Infections , Vagina/metabolism , Adolescent , Biomarkers , Burkina Faso , Cohort Studies , Female , Humans , Lactoferrin/metabolism , Randomized Controlled Trials as Topic , Reproductive Tract Infections/blood , Reproductive Tract Infections/epidemiology , Reproductive Tract Infections/metabolism , Vagina/chemistry
18.
PLoS One ; 13(4): e0195165, 2018.
Article in English | MEDLINE | ID: mdl-29624589

ABSTRACT

Chlamydia trachomatis urogenital serovars primarily replicate in epithelial cells lining the reproductive tract. Epithelial cells recognize Chlamydia through cell surface and cytosolic receptors, and/or endosomal innate receptors such as Toll-like receptors (TLRs). Activation of these receptors triggers both innate and adaptive immune mechanisms that are required for chlamydial clearance, but are also responsible for the immunopathology in the reproductive tract. We previously demonstrated that Chlamydia muridarum (Cm) induces IFN-ß in oviduct epithelial cells (OE) in a TLR3-dependent manner, and that the synthesis of several cytokines and chemokines are diminished in Cm-challenged OE derived from TLR3-/- 129S1 mice. Furthermore, our in vitro studies showed that Cm replication in TLR3-/- OE is more efficient than in wild-type OE. Because TLR3 modulates the release inflammatory mediators involved in host defense during Cm infection, we hypothesized that TLR3 plays a protective role against Cm-induced genital tract pathology in congenic C57BL/6N mice. Using the Cm mouse model for human Chlamydia genital tract infections, we demonstrated that TLR3-/- mice had increased Cm shedding during early and mid-stage genital infection. In early stage infection, TLR3-/- mice showed a diminished synthesis of IFN-ß, IL-1ß, and IL-6, but enhanced production of IL-10, TNF-α, and IFN-γ. In mid-stage infection, TLR3-/- mice exhibited significantly enhanced lymphocytic endometritis and salpingitis than wild-type mice. These lymphocytes were predominantly scattered along the endometrial stroma and the associated smooth muscle, and the lamina propria supporting the oviducts. Surprisingly, our data show that CD4+ T-cells are significantly enhanced in the genital tract TLR3-/- mice during mid-stage Chlamydial infection. In late-stage infections, both mouse strains developed hydrosalpinx; however, the extent of hydrosalpinx was more severe in TLR3-/- mice. Together, these data suggest that TLR3 promotes the clearance of Cm during early and mid-stages of genital tract infection, and that loss of TLR3 is detrimental in the development hydrosalpinx.


Subject(s)
Chlamydia Infections/genetics , Chlamydia Infections/microbiology , Chlamydia muridarum , Disease Susceptibility , Reproductive Tract Infections/genetics , Reproductive Tract Infections/microbiology , Toll-Like Receptor 3/genetics , Animals , Chlamydia Infections/metabolism , Chlamydia Infections/pathology , Cytokines/metabolism , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Reproductive Tract Infections/metabolism , Reproductive Tract Infections/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Toll-Like Receptor 3/metabolism
19.
Sci Rep ; 6: 26836, 2016 05 31.
Article in English | MEDLINE | ID: mdl-27241677

ABSTRACT

Postpartum women are at increased risk of developing puerperal sepsis caused by group A Streptococcus (GAS). Specific GAS serotypes, including M1 and M28, are more commonly associated with puerperal sepsis. However, the mechanisms of GAS genital tract infection are not well understood. We utilized a murine genital tract carriage model to demonstrate that M1 and M28 GAS colonization triggers TNF-α, IL-1ß, and IL-17A production in the female genital tract. GAS-induced IL-17A significantly influences streptococcal carriage and alters local inflammatory responses in two genetically distinct inbred strains of mice. An absence of IL-17A or the IL-1 receptor was associated with reduced neutrophil recruitment to the site of infection; and clearance of GAS was significantly attenuated in IL-17A(-/-) mice and Rag1(-/-) mice (that lack mature lymphocytes) but not in mice deficient for the IL-1 receptor. Together, these findings support a role for IL-17A in contributing to the control of streptococcal mucosal colonization and provide new insight into the inflammatory mediators regulating host-pathogen interactions in the female genital tract.


Subject(s)
Inflammation/immunology , Interleukin-17/immunology , Reproductive Tract Infections/immunology , Reproductive Tract Infections/microbiology , Streptococcal Infections/immunology , Streptococcus pyogenes/immunology , Animals , Female , Inflammation/metabolism , Inflammation/microbiology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Lymphocytes/immunology , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Reproductive Tract Infections/metabolism , Vagina/immunology , Vagina/metabolism , Vagina/microbiology
20.
PLoS One ; 9(4): e95076, 2014.
Article in English | MEDLINE | ID: mdl-24736397

ABSTRACT

The female lower genital tract is constantly exposed to microbial infection, some of which can ascend to and cause pathology such as hydrosalpinx in the upper genital tract, which can affect fertility. To understand host mechanisms for preventing upper genital tract pathology, we screened 11 inbred strains of mice for hydrosalpinx induction by C. muridarum. When examined on days 60 to 80 after intravaginal infection, the 11 strains fell into 3 groups based on their hydrosalpinx severity: CBA/J and SJL/J mice were highly susceptible with a hydrosalpinx score of 5 or greater; Balb/c, C57BL/6J, C57BL/10J, C3H/HeJ and C3H/HeN were susceptible with a score between 1 and <5; NOD/ShiLtJ, DBA/1J, DBA/2J and A/J were resistant with a score of <1. The diverse range of mouse susceptibility to hydrosalpinx induction may reflect the varied clinical outcomes of C. trachomatis-infected women. When the 11 strains were infected via an intrauterine inoculation to bypass the requirement for ascension, higher incidence and more severe hydrosalpinges were induced in most mice, indicating that the interaction between chlamydial ascension and host control of ascension is critical for determining susceptibility to hydrosalpinx development in many mice. However, a few mouse strains resisted significant exacerbation of hydrosalpinx by intrauterine infection, indicating that these mice have evolved ascension-independent mechanisms for preventing upper genital tract pathology. Together, the above observations have demonstrated that different strains of mice can prevent upper genital tract pathology by using different mechanisms.


Subject(s)
Chlamydia Infections/microbiology , Chlamydia muridarum , Host-Pathogen Interactions , Reproductive Tract Infections/microbiology , Reproductive Tract Infections/pathology , Animals , Cytokines/metabolism , Disease Models, Animal , Female , H-2 Antigens/genetics , Haplotypes , Mice , Reproductive Tract Infections/genetics , Reproductive Tract Infections/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL