1
|
Morselli S, Ceccarani C, Djusse ME, Laghi L, Camboni T, Consolandi C, Foschi C, Severgnini M, Marangoni A. Anti-chlamydial activity of vaginal fluids: new evidence from an in vitro model. Front Cell Infect Microbiol 2024; 14:1403782. [PMID: 38912205 PMCID: PMC11193362 DOI: 10.3389/fcimb.2024.1403782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/01/2024] [Indexed: 06/25/2024] Open
Abstract
Introduction We assessed the in vitro anti-chlamydial activity of fresh vaginal secretions, deciphering the microbial and metabolic components able to counteract Chlamydia trachomatis viability. Methods Forty vaginal samples were collected from a group of reproductive-aged women and their anti-chlamydial activity was evaluated by inhibition experiments. Each sample underwent 16S rRNA metabarcoding sequencing to determine the bacterial composition, as well as 1H-NMR spectroscopy to detect and quantify the presence of vaginal metabolites. Results Samples characterized by a high anti-chlamydial activity were enriched in Lactobacillus, especially Lactobacillus crispatus and Lactobacillus iners, while not-active samples exhibited a significant reduction of lactobacilli, along with higher relative abundances of Streptococcus and Olegusella. Lactobacillus gasseri showed an opposite behavior compared to L. crispatus, being more prevalent in not-active vaginal samples. Higher concentrations of several amino acids (i.e., isoleucine, leucine, and aspartate; positively correlated to the abundance of L. crispatus and L. jensenii) lactate, and 4-aminobutyrate were the most significant metabolic fingerprints of highly active samples. Acetate and formate concentrations, on the other hand, were related to the abundances of a group of anaerobic opportunistic bacteria (including Prevotella, Dialister, Olegusella, Peptostreptococcus, Peptoniphilus, Finegoldia and Anaerococcus). Finally, glucose, correlated to Streptococcus, Lachnospira and Alloscardovia genera, emerged as a key molecule of the vaginal environment: indeed, the anti-chlamydial effect of vaginal fluids decreased as glucose concentrations increased. Discussion These findings could pave the way for novel strategies in the prevention and treatment of chlamydial urogenital infections, such as lactobacilli probiotic formulations or lactobacilli-derived postbiotics.
Collapse
Affiliation(s)
- Sara Morselli
- Section of Microbiology, Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Camilla Ceccarani
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
- National Biodiversity Future Center S.c.a.r.l., Palermo, Italy
| | - Marielle Ezekielle Djusse
- Section of Microbiology, Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| | - Luca Laghi
- Department of Agricultural and Food Sciences, University of Bologna, Cesena, Italy
| | - Tania Camboni
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
| | - Clarissa Consolandi
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
- National Biodiversity Future Center S.c.a.r.l., Palermo, Italy
| | - Claudio Foschi
- Section of Microbiology, Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Marco Severgnini
- Institute of Biomedical Technologies, National Research Council, Segrate, Italy
- National Biodiversity Future Center S.c.a.r.l., Palermo, Italy
| | - Antonella Marangoni
- Section of Microbiology, Department of Medical and Surgical Sciences, Alma Mater Studiorum - University of Bologna, Bologna, Italy
| |
Collapse
|
2
|
Kamel M, Aleya S, Alsubih M, Aleya L. Microbiome Dynamics: A Paradigm Shift in Combatting Infectious Diseases. J Pers Med 2024; 14:217. [PMID: 38392650 PMCID: PMC10890469 DOI: 10.3390/jpm14020217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/24/2024] Open
Abstract
Infectious diseases have long posed a significant threat to global health and require constant innovation in treatment approaches. However, recent groundbreaking research has shed light on a previously overlooked player in the pathogenesis of disease-the human microbiome. This review article addresses the intricate relationship between the microbiome and infectious diseases and unravels its role as a crucial mediator of host-pathogen interactions. We explore the remarkable potential of harnessing this dynamic ecosystem to develop innovative treatment strategies that could revolutionize the management of infectious diseases. By exploring the latest advances and emerging trends, this review aims to provide a new perspective on combating infectious diseases by targeting the microbiome.
Collapse
Affiliation(s)
- Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza 11221, Egypt
| | - Sami Aleya
- Faculty of Medecine, Université de Bourgogne Franche-Comté, Hauts-du-Chazal, 25030 Besançon, France;
| | - Majed Alsubih
- Department of Civil Engineering, King Khalid University, Guraiger, Abha 62529, Saudi Arabia;
| | - Lotfi Aleya
- Laboratoire de Chrono-Environnement, Université de Bourgogne Franche-Comté, UMR CNRS 6249, La Bouloie, 25030 Besançon, France;
| |
Collapse
|
3
|
Zou X, Nakura Y, Kawaguchi H, Nishiumi F, Wu HN, Yanagihara I. Comparison of databases useful for the analysis of vaginal microbiota in Japanese women using next-generation sequencing data (QIIME 2 software). J Appl Microbiol 2023; 134:lxad283. [PMID: 38012110 DOI: 10.1093/jambio/lxad283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 11/17/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023]
Abstract
AIMS Approximately 10% of children are born prematurely, and bacterial vaginosis during pregnancy is associated with preterm delivery. Highly accurate species-level vaginal microflora analysis helps control bacteria-induced preterm birth. Therefore, we aimed to conduct a bioinformatic analysis of gene sequences using 16S databases and compare their efficacy in comprehensively identifying potentially pathogenic vaginal microbiota in Japanese women. METHODS AND RESULTS The 16 s rRNA databases, Silva, Greengenes, and the basic local alignment search tool (BLAST) were compared to determine whether the classification quality could be improved using the V3-V4 region next-generation sequencing (NGS) sequences. It was found that NGS data were aligned using the BLAST database with the QIIME 2 platform, whose classification quality was higher than that of Silva, and the combined Silva and Greengenes databases based on the mutual complementarity of the two databases. CONCLUSIONS The reference database selected during the bioinformatic processing influenced the recognized sequence percentage, taxonomic rankings, and accuracy. This study showed that the BLAST database was the best choice for NGS data analysis of Japanese women's vaginal microbiota.
Collapse
Affiliation(s)
- Xianya Zou
- Department of Developmental Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi City, Osaka 594-1101, Japan
- Department of Pediatric and Neonatal-Perinatal Research, Graduate School of Medicine, Osaka University, 1-1 Yamadaoka, Suita City, Osaka 565-0871, Japan
| | - Yukiko Nakura
- Department of Developmental Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi City, Osaka 594-1101, Japan
| | - Haruna Kawaguchi
- Department of Developmental Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi City, Osaka 594-1101, Japan
- Department of Obstetrics, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi City, Osaka 594-1101, Japan
| | - Fumiko Nishiumi
- Department of Developmental Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi City, Osaka 594-1101, Japan
| | - Heng Ning Wu
- Department of Developmental Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi City, Osaka 594-1101, Japan
| | - Itaru Yanagihara
- Department of Developmental Medicine, Research Institute, Osaka Women's and Children's Hospital, 840 Murodo-cho, Izumi City, Osaka 594-1101, Japan
| |
Collapse
|
4
|
Savicheva AM, Krysanova AA, Budilovskaya OV, Spasibova EV, Khusnutdinova TA, Shalepo KV, Beliaeva NR, Safarian GK, Sapozhnikov KV, Tapilskaya NI, Kogan IY. Vaginal Microbiota Molecular Profiling in Women with Bacterial Vaginosis: A Novel Diagnostic Tool. Int J Mol Sci 2023; 24:15880. [PMID: 37958862 PMCID: PMC10649576 DOI: 10.3390/ijms242115880] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/27/2023] [Accepted: 10/31/2023] [Indexed: 11/15/2023] Open
Abstract
Bacterial vaginosis (BV) is a most common microbiological syndrome. Multiplex next-generation sequencing (NGS) or molecular tests allow a complete and accurate vaginal microbiota profiling in order to determine the primary causative agent. Due to the high costs and limited availability of NGS, the multiplex real-time PCR draws more attention. The present study aimed to evaluate the microbial composition and dominant lactobacilli species in non-pregnant women with bacterial vaginosis using a multiplex RT-PCR test and determine its diagnostic significance. In total, 331 women complaining of vaginal discharge were included. BV was confirmed upon clinical examination and Nugent criteria. A real-time PCR test was carried out with a new Femoflor test, which identifies opportunistic bacteria, STD pathogens, and some viruses. According to the results, the rate of lactobacilli is significantly reduced in BV-affected patients when compared to healthy women. Moreover, the rate of L. crispatus significantly decreases, while the rate of L. iners remains high. Among obligate anaerobic bacteria, Gardnerella vaginalis was the most prevalent in women with BV. The Femoflor test demonstrated high sensitivity and specificity for diagnosing BV. Moreover, the test allows the identification of infection in women with intermediate vaginal microbiota, as well as STD pathogens, and viruses. Thus, the application of real-time PCR tests can be effectively used in vaginal microbiota evaluation in women with BV, intermediate vaginal microbiota, and healthy women. In addition, this test may be used as an alternative to the Amsel criteria and Nugent scoring method in diagnosing BV.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Galina Kh. Safarian
- D.O. Ott Research Institute of Obstetrics, Gynecology and Reproductive Medicine, 199034 St. Petersburg, Russia; (A.M.S.); (A.A.K.); (O.V.B.); (E.V.S.); (T.A.K.); (K.V.S.); (N.R.B.); (K.V.S.); (N.I.T.); (I.Y.K.)
| | | | | | | |
Collapse
|
5
|
Banerjee P, Gaddam N, Chandler V, Chakraborty S. Oxidative Stress-Induced Liver Damage and Remodeling of the Liver Vasculature. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1400-1414. [PMID: 37355037 DOI: 10.1016/j.ajpath.2023.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/29/2023] [Accepted: 06/08/2023] [Indexed: 06/26/2023]
Abstract
As an organ critically important for targeting and clearing viruses, bacteria, and other foreign material, the liver operates via immune-tolerant, anti-inflammatory mechanisms indispensable to the immune response. Stress and stress-induced factors disrupt the homeostatic balance in the liver, inflicting tissue damage, injury, and remodeling. These factors include oxidative stress (OS) induced by viral infections, environmental toxins, drugs, alcohol, and diet. A recurrent theme seen among stressors common to multiple liver diseases is the induction of mitochondrial dysfunction, increased reactive oxygen species expression, and depletion of ATP. Inflammatory signaling additionally exacerbates the condition, generating a proinflammatory, immunosuppressive microenvironment and activation of apoptotic and necrotic mechanisms that disrupt the integrity of liver morphology. These pathways initiate signaling pathways that significantly contribute to the development of liver steatosis, inflammation, fibrosis, cirrhosis, and liver cancers. In addition, hypoxia and OS directly enhance angiogenesis and lymphangiogenesis in chronic liver diseases. Late-stage consequences of these conditions often narrow the outcomes for liver transplantation or result in death. This review provides a detailed perspective on various stress-induced factors and the specific focus on role of OS in different liver diseases with special emphasis on different molecular mechanisms. It also highlights how resultant changes in the liver vasculature correlate with pathogenesis.
Collapse
Affiliation(s)
- Priyanka Banerjee
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, Texas.
| | - Niyanshi Gaddam
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, Texas
| | - Vanessa Chandler
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, Texas
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M Health Science Center, Bryan, Texas.
| |
Collapse
|
6
|
Zalambani C, Rizzardi N, Marziali G, Foschi C, Morselli S, Djusse ME, Naldi M, Fato R, Calonghi N, Marangoni A. Role of D(-)-Lactic Acid in Prevention of Chlamydia trachomatis Infection in an In Vitro Model of HeLa Cells. Pathogens 2023; 12:883. [PMID: 37513730 PMCID: PMC10383594 DOI: 10.3390/pathogens12070883] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023] Open
Abstract
A vaginal microbiota dominated by certain Lactobacillus species may have a protective effect against Chlamydia trachomatis infection. One of the key antimicrobial compounds produced is lactic acid, which is believed to play a central role in host defense. Lactobacillus strains producing the D(-)-lactic acid isomer are known to exert stronger protection. However, the molecular mechanisms underlying this antimicrobial action are not well understood. The aim of this study was to investigate the role of D(-)-lactic acid isomer in the prevention of C. trachomatis infection in an in vitro HeLa cell model. We selected two strains of lactobacilli belonging to different species: a vaginal isolate of Lactobacillus crispatus that releases both D(-) and L(+) isomers and a strain of Lactobacillus reuteri that produces only the L(+) isomer. Initially, we demonstrated that L. crispatus was significantly more effective than L. reuteri in reducing C. trachomatis infectivity. A different pattern of histone acetylation and lactylation was observed when HeLa cells were pretreated for 24 h with supernatants of Lactobacillus crispatus or L. reuteri, resulting in different transcription of genes such as CCND1, CDKN1A, ITAG5 and HER-1. Similarly, distinct transcription patterns were found in HeLa cells treated with 10 mM D(-)- or L(+)-lactic acid isomers. Our findings suggest that D(-) lactic acid significantly affects two non-exclusive mechanisms involved in C. trachomatis infection: regulation of the cell cycle and expression of EGFR and α5β1-integrin.
Collapse
Affiliation(s)
- Chiara Zalambani
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Nicola Rizzardi
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Giacomo Marziali
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Claudio Foschi
- Microbiology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Sara Morselli
- Microbiology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Marielle Ezekielle Djusse
- Microbiology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Marina Naldi
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Romana Fato
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Natalia Calonghi
- Department of Pharmacy and Biotechnology (FABIT), University of Bologna, 40126 Bologna, Italy
| | - Antonella Marangoni
- Microbiology, Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
7
|
Heczko P, Kozień Ł, Strus M. Special Issue "An Update on Lactobacillus": Editorial. Microorganisms 2023; 11:1400. [PMID: 37374902 DOI: 10.3390/microorganisms11061400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/19/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
As indicated in the introduction to this Special Issue, as of 2020, the original genus Lactobacillus comprised over 260 recognized species, a figure which is probably much higher now [...].
Collapse
Affiliation(s)
- Piotr Heczko
- Chair of Microbiology, Department of Bacteriology, Microbial Ecology and Parasitology, Jagiellonian University Medical College, 31-121 Cracow, Poland
| | - Łucja Kozień
- Chair of Microbiology, Department of Bacteriology, Microbial Ecology and Parasitology, Jagiellonian University Medical College, 31-121 Cracow, Poland
| | - Magdalena Strus
- Chair of Microbiology, Department of Bacteriology, Microbial Ecology and Parasitology, Jagiellonian University Medical College, 31-121 Cracow, Poland
| |
Collapse
|
8
|
Liu P, Lu Y, Li R, Chen X. Use of probiotic lactobacilli in the treatment of vaginal infections: In vitro and in vivo investigations. Front Cell Infect Microbiol 2023; 13:1153894. [PMID: 37077531 PMCID: PMC10106725 DOI: 10.3389/fcimb.2023.1153894] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/23/2023] [Indexed: 04/05/2023] Open
Abstract
The vaginal microbiome is a distinct component of the human microbiome that is colonized by a wide variety of microorganisms. Lactobacilli are the most frequently identified microorganisms in the healthy human vagina. These Gram-positive bacilli can acidify the vaginal microenvironment, inhibit the proliferation of other pathogenic microorganisms, and promote the maintenance of a eubiotic vaginal microbiome. However, a vaginal flora with a reduced proportion or abundance of lactobacilli is associated with various vaginal infections that have been linked to serious health consequences such as infertility, preterm birth, pelvic inflammatory disease, premature rupture of membranes, and miscarriage. Due to their “Generally Recognized as Safe” classification and critical role in vaginal health, probiotic lactobacilli have been widely used as an alternative or adjunct to traditional antibiotic therapy for the treatment of vaginal infections and restoration of the vaginal microbiome. This review focuses on the significant role of probiotic lactobacilli in the vaginal microenvironment and discusses the use of probiotic lactobacilli in the treatment of female vaginal infections in vitro and in vivo.
Collapse
Affiliation(s)
| | | | - Rongguo Li
- *Correspondence: Rongguo Li, ; Xiaodi Chen,
| | | |
Collapse
|
9
|
Giordani B, Naldi M, Croatti V, Parolin C, Erdoğan Ü, Bartolini M, Vitali B. Exopolysaccharides from vaginal lactobacilli modulate microbial biofilms. Microb Cell Fact 2023; 22:45. [PMID: 36890519 PMCID: PMC9993704 DOI: 10.1186/s12934-023-02053-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 03/02/2023] [Indexed: 03/10/2023] Open
Abstract
BACKGROUND Exopolysaccharides (EPS) secreted by beneficial lactobacilli exert a plethora of positive activities, but little is known about their effects on biofilms of opportunistic vaginal pathogens and especially on biofilms of lactobacilli themselves. Here, the EPS produced by six vaginal lactobacilli, belonging to Lactobacillus crispatus (BC1, BC4, BC5) and Lactobacillus gasseri (BC9, BC12, BC14) species were isolated from cultural supernatants and lyophilized. RESULTS Lactobacillus EPS were chemically characterized in terms of monosaccharide composition by liquid chromatography (LC) analysis coupled to UV and mass spectrometry (MS) detection. Moreover, the ability of EPS (0.1, 0.5, 1 mg/mL) to stimulate the biofilm formation of lactobacilli and to inhibit the formation of pathogens' biofilms was evaluated by crystal violet (CV) staining and 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay. Isolated EPS (yields 133-426 mg/L) were heteropolysaccharides mainly composed of D-mannose (40-52%) and D-glucose (11-30%). For the first time we demonstrated that Lactobacillus EPS were able to stimulate in a dose-dependent manner (p < 0.05) the formation of biofilms of ten strains belonging to L. crispatus, L. gasseri and Limosilactobacillus vaginalis species, in terms of cell viability (84-282% increase at 1 mg/mL) and especially biofilm biomass (40-195% increase at 1 mg/mL), quantified with MTT assay and CV staining, respectively. EPS released from L. crispatus and L. gasseri were found to better stimulate the biofilms of the same producer species rather than that of other species, including producing strains themselves and other strains. Conversely, the biofilm formation of bacterial (Escherichia coli, Staphylococcus spp., Enterococcus spp. and Streptococcus agalactiae) and fungal (Candida spp.) pathogens was inhibited. The anti-biofilm activity was dose-dependent and was more marked for L. gasseri-derived EPS (inhibition up to 86%, 70%, and 58% at 1 mg/mL, 0.5 mg/mL, and 0.1 mg/mL, respectively), whilst L. crispatus-derived EPS resulted overall less efficient (inhibition up to 58% at 1 mg/mL and 40% at 0.5 mg/mL) (p < 0.05). CONCLUSIONS Lactobacilli-derived EPS favour the biofilm formation of lactobacilli preventing, at the same time, that of opportunistic pathogens. These results support the possible employment of EPS as postbiotics in medicine as a therapeutic/preventive strategy to counteract vaginal infections.
Collapse
Affiliation(s)
- Barbara Giordani
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Marina Naldi
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Vanessa Croatti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Carola Parolin
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | | | - Manuela Bartolini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Beatrice Vitali
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.
| |
Collapse
|
10
|
D'Alessandro M, Gottardi D, Parolin C, Glicerina VT, Vitali B, Lanciotti R, Patrignani F. Development and characterization of fermented soy milks containing encapsulated or non-encapsulated vaginal probiotics. Lebensm Wiss Technol 2023. [DOI: 10.1016/j.lwt.2023.114713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
|
11
|
Lu B, Qiao Q, Park ER, Wang Y, Gilleran JA, Pan M, Pilch DS, Wu X, Roberge JY, Fan H. Acylpyrazoline-Based Third-Generation Selective Antichlamydial Compounds with Enhanced Potency. ACS OMEGA 2023; 8:6597-6607. [PMID: 36844602 PMCID: PMC9947980 DOI: 10.1021/acsomega.2c06992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/30/2023] [Indexed: 06/18/2023]
Abstract
Chlamydiae are obligate intracellular Gram-negative bacteria and widespread pathogens in humans and animals. Broad-spectrum antibiotics are currently used to treat chlamydial infections. However, broad-spectrum drugs also kill beneficial bacteria. Recently, two generations of benzal acylhydrazones have been shown to selectively inhibit chlamydiae without toxicity to human cells and lactobacilli, which are dominating, beneficial bacteria in the vagina of reproductive-age women. Here, we report the identification of two acylpyrazoline-based third-generation selective antichlamydials (SACs). With minimal inhibitory concentrations (MIC) and minimal bactericidal concentrations (MBC) of 10-25 μM against Chlamydia trachomatis and Chlamydia muridarum, these new antichlamydials are 2- to 5-fold more potent over the benzal acylhydrazone-based second-generation selective antichlamydial lead SF3. Both acylpyrazoline-based SACs are well tolerated by Lactobacillus, Escherichia coli, Klebsiella, and Salmonella as well as host cells. These third-generation selective antichlamydials merit further evaluation for therapeutic application.
Collapse
Affiliation(s)
- Bin Lu
- Department
of Parasitology, Central South University
Xiangya Medical School, Changsha, Hunan 410013, China
- Department
of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Qi Qiao
- Molecular
Design and Synthesis Core, RUBRIC, Office for Research, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Elizabeth R. Park
- Molecular
Design and Synthesis Core, RUBRIC, Office for Research, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
- Department
of Chemistry and Chemical Biology, Rutgers,
The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Yuxuan Wang
- Department
of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - John A. Gilleran
- Molecular
Design and Synthesis Core, RUBRIC, Office for Research, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Matthew Pan
- Department
of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Daniel S. Pilch
- Department
of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Xiang Wu
- Department
of Parasitology, Central South University
Xiangya Medical School, Changsha, Hunan 410013, China
| | - Jacques Y. Roberge
- Molecular
Design and Synthesis Core, RUBRIC, Office for Research, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| | - Huizhou Fan
- Department
of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, New Jersey 08854, United States
| |
Collapse
|
12
|
Giordani B, Abruzzo A, Parolin C, Foschi C, Laghi L, Marangoni A, Luppi B, Vitali B. Prebiotic Activity of Vaginal Lactobacilli on Bifidobacteria: from Concept to Formulation. Microbiol Spectr 2023; 11:e0200922. [PMID: 36602371 PMCID: PMC9927276 DOI: 10.1128/spectrum.02009-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The gut of babies born vaginally is rapidly colonized by Bifidobacterium spp. after birth, while in infants born by cesarean section (C-section), the presence of bifidobacteria drops dramatically, increasing the risk of developing gastrointestinal disorders. Considering that newborns naturally come into contact with maternal lactobacilli as they pass through the birth canal, the aim of this work is to exploit for the first time the bifidogenic activity exerted by the cell-free supernatants (CFSs) from lactobacilli of vaginal origin, belonging to the species Lactobacillus crispatus, Lactobacillus gasseri, Limosilactobacillus vaginalis, and Lactiplantibacillus plantarum. CFSs were recovered after 7 h, 13 h, and 24 h of fermentation and assessed for the ability to stimulate the planktonic growth and biofilms of Bifidobacterium strains belonging to species widely represented in the gut tract. A bifidogenic effect was observed for all CFSs; such activity was maximal for CFSs recovered in exponential phase and was strongly dependent on the species of lactobacilli. Importantly, no stimulating effects on an intestinal Escherichia coli strain were observed. CFSs from L. vaginalis BC17 showed the best bifidogenic profile since they increased bifidobacterial planktonic growth by up to 432% and biofilm formation by up to 289%. The CFS at 7 h from BC17 was successfully formulated with a hyaluronic acid-based hydrogel aimed at preventing and treating breast sores in lactating women and exerting bifidogenic activity in infants born mainly by C-section. IMPORTANCE Bifidobacteria in the gut tract of infants play crucial roles in the prevention of gastrointestinal diseases and the maturation of the immune system. Consequently, strategies to trigger a bifidogenic shift in the infant gut are highly desirable. Evidences suggest that the presence of a maternal vaginal microbiota dominated by health-promoting lactobacilli and the development of a bifidobacterium-enriched gut microbiota in newborns are interconnected. In this context, we found out that the cell-free supernatants from lactobacilli of vaginal origin were able to effectively stimulate the proliferation of Bifidobacterium spp. grown in free-floating and biofilm forms. The cell-free supernatant from Limosilactobacillus vaginalis BC17 showed excellent bifidogenic behavior, which was preserved even after its incorporation into a nipple formulation for lactating women. Lactobacilli derivatives, such as cell-free supernatants, have gained increasing interest by virtue of their safer profile than that of living cells and can be proposed as an ecosustainable approach to favor gut colonization of infants by bifidobacteria.
Collapse
Affiliation(s)
- Barbara Giordani
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Angela Abruzzo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Carola Parolin
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Claudio Foschi
- Section of Microbiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
- Microbiology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Luca Laghi
- Department of Agricultural and Food Sciences, University of Bologna, Cesena, Italy
| | - Antonella Marangoni
- Section of Microbiology, Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Barbara Luppi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Beatrice Vitali
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
13
|
Xiao Y, Huang S, Yu W, Ni Y, Lu D, Wu Q, Leng Q, Yang T, Ni M, Xie J, Zhang X. Effects of emergency/nonemergency cervical cerclage on the vaginal microbiome of pregnant women with cervical incompetence. Front Cell Infect Microbiol 2023; 13:1072960. [PMID: 36968117 PMCID: PMC10034410 DOI: 10.3389/fcimb.2023.1072960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/21/2023] [Indexed: 03/29/2023] Open
Abstract
Background Evaluation of the therapeutic effects of cerclage on preterm birth (PTB) caused by cervical incompetence remains challenging. The vaginal microbiome is associated with preterm births. Thus, this study aimed to analyse the vaginal microbiota of patients with cervical incompetence, explore the relationship between the composition of the vaginal microbiota before cervical cerclage and at term delivery, and assess the effect of cervical cerclage on the vaginal microbiota. Methods Patients (n = 30) underwent cerclage performed by the same surgical team. Vaginal swabs were obtained pre-surgery and seven days post-surgery. A gestational age-matched cohort of healthy pregnant women (n = 20) (no particular abnormality during pregnancy, delivery at term) was used as the control group and sampled during a comparable pregnancy. All collected vaginal swabs were analysed by 16S rRNA gene sequencing. Results When comparing the healthy control and cervical cerclage groups, the enriched microorganism in the healthy controls was G. Scardovia, and the enriched microorganism of the cerclage was G. Streptococcus. α diversity was significantly increased in patients who received cerclage with preterm delivery compared with those with full-term delivery, and the enriched microorganism was F. Enterococcus. A comparison before and after nonemergency cerclage suggested that the enriched microorganisms were G. Lactobacillus and F. Lactobacillaceae before surgery. After nonemergency cerclage, the enriched microorganisms were F. Enterobacteriaceae and C. Gammaproteobacteria. Vaginal microbiota diversity significantly increased, and the proportion of women with Lactobacillus spp.-depleted microbiomes increased after emergency cerclage. Significant differences in β diversity were found between the groups. Before the emergency cerclage, the enriched microorganisms were G. Lactobacillus, O. Alteromonadales, and P. Firmicutes. After emergency cerclage, the enriched microorganisms were P. Actinobacteria, C. Actinobacteria, P. Proteobacteria, F. Bifidobacteriaceae, O. Bifidobacteriales, G. Gardnerella, and G. Veillonella. Conclusion Cerclage (particularly emergency cerclage) may alter the vaginal microbiota by increasing microbiota diversity, decreasing vaginal Lactobacillus abundance, and increasing the abundance of pathogenic bacteria that are not conducive to pregnancy maintenance, thereby affecting surgical efficacy. Therefore, the role of the vaginal microbiome should be considered when developing treatment strategies for pregnant women with cervical incompetence. Clinical trial registration https://www.chictr.org.cn, identifier ChiCTR2100046305.
Collapse
Affiliation(s)
- Yunshan Xiao
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Xiamen, China
- Xiamen Clinical Research Center for Perinatal Medicine, Xiamen, China
| | - Shiting Huang
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Xiamen, China
| | - Weiwei Yu
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Xiamen, China
| | - Yan Ni
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Xiamen, China
| | - Danni Lu
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Xiamen, China
| | - Quanfeng Wu
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Xiamen, China
| | - Qin Leng
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Xiamen, China
| | - Ting Yang
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Xiamen, China
| | - Meilan Ni
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Xiamen, China
| | - Jingxian Xie
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Xiamen, China
| | - Xueqin Zhang
- Department of Obstetrics, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Basic and Clinical Research on Major Obstetrical Diseases, Xiamen, China
- Xiamen Clinical Research Center for Perinatal Medicine, Xiamen, China
- *Correspondence: Xueqin Zhang,
| |
Collapse
|
14
|
Sun Z, Ge X, Qiu B, Xiang Z, Jiang C, Wu J, Li Y. Vulvovaginal candidiasis and vaginal microflora interaction: Microflora changes and probiotic therapy. Front Cell Infect Microbiol 2023; 13:1123026. [PMID: 36816582 PMCID: PMC9936092 DOI: 10.3389/fcimb.2023.1123026] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
Vaginal microbiome is mutually beneficial to the host and has a significant impact on health and disease. Candida species, including Candida albicans, are part of the mucosal flora of most healthy women. Under suitable conditions, they can live in the vulvovaginal mucosa, resulting in symptomatic vulvovaginal candidiasis (VVC). Based on the analysis of 16S ribosomal RNA gene sequences, great progress has been made in exploring the composition and structure of vaginal bacterial community. Moreover, researchers have conducted several studies on whether vaginal microbiome will change during VVC infection. In addition, it has been reported that vaginal colonization of probiotics in vaginal microorganisms, especially Lactobacillus, can effectively reduce the risk of VVC and treat VVC. This review aims to summarize the changes of vaginal microflora during VVC infection, and further point out the possibility of using lactic acid bacteria as probiotics to treat VVC, so as to reduce the adverse consequences of VVC infection and reduce the expensive treatment cost.
Collapse
Affiliation(s)
- Zhongwen Sun
- Department of Medical Technology, Suzhou Vocational Health College, Suzhou, Jiangsu, China
| | - Xinnuo Ge
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bo Qiu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ze Xiang
- Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chun Jiang
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Jian Wu
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| | - Yuan Li
- Departments of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China
| |
Collapse
|
15
|
Han X, Du S, Chen X, Min X, Dong Z, Wang Y, Zhu C, Wei F, Gao S, Cai Q. Lactate-mediated Fascin protrusions promote cell adhesion and migration in cervical cancer. Theranostics 2023; 13:2368-2383. [PMID: 37153736 PMCID: PMC10157738 DOI: 10.7150/thno.83938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 03/13/2023] [Indexed: 05/10/2023] Open
Abstract
Background: Lactate is associated with the poor prognosis of many human malignancies. Cervical cancer, one of main causes of women mortality worldwide, is aggressive and absent of effective pharmacological treatment, and its underlying mechanisms of progression remain elusive. Methods: The regulation of β-catenin to fascin protrusion formation upon acidic lactate (Lactic acid [LA]) stimulation was evaluated through in β-catenin or fascin deficiency cell line models by immunofluorescence assays, and subcellular fractionation. The effect of β-catenin and fascin relocation by LA and its antagonist were evaluated by immunohistochemistry assay in patient tissues and mouse tumor xenograft model. Trypsin digestion, Transwell assay, cell proliferation in vitro was performed to explore the role of LA in the cell growth, adhesion and migration. Results: Low concentration of LA significantly promotes cytoskeleton remodeling via `protrusion formation to increase cell adhesion and migration. Mechanistically, upon LA stimulation, β-catenin diffuses from the cytoplasmic membrane into the nucleus, which in turn induces fascin nuclear-cytoplasm redistribution to the protrusion compartment. Moreover, the antagonist of LA sufficiently blocks the LA-mediated β-catenin nuclear import, fascin nuclear export, and the growth and invasion of cervical cancer cells in vitro and in vivo using a murine xenograft model. Conclusions: This study uncovers β-catenin-fascin axis as a key signal in response to extracellular lactate and indicates that antagonist of LA may serve as a potential clinical intervention for cancer development.
Collapse
Affiliation(s)
- Xiao Han
- Center of Diagnosis and Treatment For Cervical & Uterine Cavity Disease, Obstetrics and Gynecology Hospital of Fudan University, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, & MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Shujuan Du
- Center of Diagnosis and Treatment For Cervical & Uterine Cavity Disease, Obstetrics and Gynecology Hospital of Fudan University, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, & MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Xiaoting Chen
- Center of Diagnosis and Treatment For Cervical & Uterine Cavity Disease, Obstetrics and Gynecology Hospital of Fudan University, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, & MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Xuehua Min
- Center of Diagnosis and Treatment For Cervical & Uterine Cavity Disease, Obstetrics and Gynecology Hospital of Fudan University, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, & MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Zhongwei Dong
- Center of Diagnosis and Treatment For Cervical & Uterine Cavity Disease, Obstetrics and Gynecology Hospital of Fudan University, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, & MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Yuyan Wang
- Center of Diagnosis and Treatment For Cervical & Uterine Cavity Disease, Obstetrics and Gynecology Hospital of Fudan University, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, & MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Caixia Zhu
- Center of Diagnosis and Treatment For Cervical & Uterine Cavity Disease, Obstetrics and Gynecology Hospital of Fudan University, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, & MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Fang Wei
- ShengYushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
- ✉ Corresponding author: (QC); (SG); (FW)
| | - Shujun Gao
- Center of Diagnosis and Treatment For Cervical & Uterine Cavity Disease, Obstetrics and Gynecology Hospital of Fudan University, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, & MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
- ✉ Corresponding author: (QC); (SG); (FW)
| | - Qiliang Cai
- Center of Diagnosis and Treatment For Cervical & Uterine Cavity Disease, Obstetrics and Gynecology Hospital of Fudan University, Shanghai Key Laboratory of Female Reproductive Endocrine-Related Disease, & MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
- ✉ Corresponding author: (QC); (SG); (FW)
| |
Collapse
|
16
|
Dong M, Dong Y, Bai J, Li H, Ma X, Li B, Wang C, Li H, Qi W, Wang Y, Fan A, Han C, Xue F. Interactions between microbiota and cervical epithelial, immune, and mucus barrier. Front Cell Infect Microbiol 2023; 13:1124591. [PMID: 36909729 PMCID: PMC9998931 DOI: 10.3389/fcimb.2023.1124591] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/31/2023] [Indexed: 02/26/2023] Open
Abstract
The female reproductive tract harbours hundreds of bacterial species and produces numerous metabolites. The uterine cervix is located between the upper and lower parts of the female genital tract. It allows sperm and birth passage and hinders the upward movement of microorganisms into a relatively sterile uterus. It is also the predicted site for sexually transmitted infection (STI), such as Chlamydia, human papilloma virus (HPV), and human immunodeficiency virus (HIV). The healthy cervicovaginal microbiota maintains cervical epithelial barrier integrity and modulates the mucosal immune system. Perturbations of the microbiota composition accompany changes in microbial metabolites that induce local inflammation, damage the cervical epithelial and immune barrier, and increase susceptibility to STI infection and relative disease progression. This review examined the intimate interactions between the cervicovaginal microbiota, relative metabolites, and the cervical epithelial-, immune-, and mucus barrier, and the potent effect of the host-microbiota interaction on specific STI infection. An improved understanding of cervicovaginal microbiota regulation on cervical microenvironment homeostasis might promote advances in diagnostic and therapeutic approaches for various STI diseases.
Collapse
Affiliation(s)
- Mengting Dong
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yalan Dong
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Junyi Bai
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huanrong Li
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaotong Ma
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Bijun Li
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Chen Wang
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Huiyang Li
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenhui Qi
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingmei Wang
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Aiping Fan
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
| | - Cha Han
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Cha Han, ; Fengxia Xue,
| | - Fengxia Xue
- Department of Obstetrics and Gynaecology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Female Reproductive Health and Eugenic, Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China
- *Correspondence: Cha Han, ; Fengxia Xue,
| |
Collapse
|
17
|
Croatti V, Parolin C, Giordani B, Foschi C, Fedi S, Vitali B. Lactobacilli extracellular vesicles: potential postbiotics to support the vaginal microbiota homeostasis. Microb Cell Fact 2022; 21:237. [PMCID: PMC9664694 DOI: 10.1186/s12934-022-01963-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 10/23/2022] [Indexed: 11/16/2022] Open
Abstract
Abstract
Background
Lactobacillus species dominate the vaginal microflora performing a first-line defense against vaginal infections. Extracellular vesicles (EVs) released by lactobacilli are considered mediators of their beneficial effects affecting cellular communication, homeostasis, microbial balance, and host immune system pathways. Up to now, very little is known about the role played by Lactobacillus EVs in the vaginal microenvironment, and mechanisms of action remain poorly understood.
Results
Here, we hypothesized that EVs can mediate lactobacilli beneficial effects to the host by modulating the vaginal microbiota colonization. We recovered and characterized EVs produced by two vaginal strains, namely Lactobacillus crispatus BC5 and Lactobacillus gasseri BC12. EVs were isolated by ultracentrifugation and physically characterized by Nanoparticle Tracking Analysis (NTA) and Dynamic Light Scattering (DLS). EVs protein and nucleic acids (DNA and RNA) content was also evaluated. We explored the role of EVs on bacterial adhesion and colonization, using a cervical cell line (HeLa) as an in vitro model. Specifically, we evaluated the effect of EVs on the adhesion of both vaginal beneficial lactobacilli and opportunistic pathogens (i.e., Escherichia coli, Staphylococcus aureus, Streptococcus agalactiae, and Enterococcus faecalis). We demonstrated that EVs from L. crispatus BC5 and L. gasseri BC12 significantly enhanced the cellular adhesion of all tested lactobacilli, reaching the maximum stimulation effect on strains belonging to L. crispatus species (335% and 269% of average adhesion, respectively). At the same time, EVs reduced the adhesion of all tested pathogens, being EVs from L. gasseri BC12 the most efficient.
Conclusions
Our observations suggest for the first time that EVs released by symbiotic Lactobacillus strains favor healthy vaginal homeostasis by supporting the colonization of beneficial species and preventing pathogens attachment. This study reinforces the concept of EVs as valid postbiotics and opens the perspective of developing postbiotics from vaginal strains to maintain microbiota homeostasis and promote women’s health.
Collapse
|
18
|
Parolin C, Croatti V, Giordani B, Vitali B. Vaginal Lactobacillus Impair Candida Dimorphic Switching and Biofilm Formation. Microorganisms 2022; 10:microorganisms10102091. [PMID: 36296367 PMCID: PMC9609122 DOI: 10.3390/microorganisms10102091] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/30/2022] [Accepted: 10/18/2022] [Indexed: 02/16/2023] Open
Abstract
Lactobacillus spp. generally dominate the vaginal microbiota and prevent pathogen adhesion and overgrowth, including Candida spp., by various mechanisms. Although Candida spp. can be commensal, in certain conditions they can become pathogenic, causing vulvovaginal candidiasis. The insurgence of candidiasis is related to the expression of Candida virulence factors, including morphologic switching and biofilm formation. Germ tubes, pseudohyphae, and hyphae promote Candida tissue invasion, biofilms increase persistence and are often resistant to antifungals and host immune response. Here, we explored the inhibitory activity of vaginal Lactobacillus strains belonging to Lactobacillus crispatus, Lactobacillus gasseri, Limosilactobacillus vaginalis, and Lactiplantibacillus plantarum species towards Candida virulence factors. With the aim to investigate the interrelation between mode of growth and functionality, supernatants were collected from lactobacilli planktonic cultures and, for the first time, from adherent ones, and were evaluated towards Candida dimorphic switching and biofilm. Candida biofilms were analyzed by multiple methodologies, i.e., crystal violet staining, MTT assay, and confocal microscopy. Lactobacillus supernatants reduce Candida switching and biofilm formation. Importantly, L. crispatus supernatants showed the best profile of virulence suppression, especially when grown in adherence. These results highlight the role of such species as a hallmark of vaginal eubiosis and prompt its employment in new probiotics for women's health.
Collapse
|
19
|
Starc M, Lučovnik M, Eržen Vrlič P, Jeverica S. Protective Effect of Lactobacillus crispatus against Vaginal Colonization with Group B Streptococci in the Third Trimester of Pregnancy. Pathogens 2022; 11:pathogens11090980. [PMID: 36145412 PMCID: PMC9506259 DOI: 10.3390/pathogens11090980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
Background: A normal vaginal microbiota may protect the vaginal mucosa from colonization by potentially pathogenic bacteria, including group B streptococci (GBS). The aim of this study was to investigate the association between colonization with GBS and the presence of specific vaginal microbiota isolated from vaginal swabs in the third trimester of pregnancy. Methods: A semiquantitative culture of 1860 vaginal swabs from consecutive pregnant women in their third trimester was analyzed. The dominant bacteria, including lactobacilli, were identified using MALDI-TOF mass spectrometry. An enrichment culture for GBS was performed on the swabs. GBS colonization correlated with the bacteria isolated at the same time. Results: Lactobacilluscrispatus was isolated in 27.5% of the cultures, followed by L. jensenii (13.9%), L. gasseri (12.6%), and L. iners (10.1%). The presence of lactobacilli as a group, and of L. crispatus, inversely correlated with GBS colonization (OR = 0.44 and OR = 0.5, respectively; both with p < 0.001). Other microorganisms, including Gardnerella vaginalis, mixed aerobic bacteria and yeasts, were not associated with GBS colonization. Conclusions: Lactobacilli, especially L. crispatus, may prevent GBS colonization in pregnancy. Maintaining a normal vaginal microbiota could be an effective method for the antibiotic-free prevention of invasive GBS infections in neonates.
Collapse
Affiliation(s)
- Maja Starc
- Department of Obstetrics and Gynecology, General Hospital Slovenj Gradec, 2380 Slovenj Gradec, Slovenia
| | - Miha Lučovnik
- Department of Perinatology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Petra Eržen Vrlič
- Department of Obstetrics and Gynecology, Community Health Center Ljubljana, 1000 Ljubljana, Slovenia
| | - Samo Jeverica
- Center for Medical Microbiology, National Laboratory of Health, Environment and Food, 2000 Maribor, Slovenia
- Correspondence: ; Tel.: +386-40-300-855
| |
Collapse
|
20
|
Chen H, Min S, Wang L, Zhao L, Luo F, Lei W, Wen Y, Luo L, Zhou Q, Peng L, Li Z. Lactobacillus Modulates Chlamydia Infectivity and Genital Tract Pathology in vitro and in vivo. Front Microbiol 2022; 13:877223. [PMID: 35572713 PMCID: PMC9098263 DOI: 10.3389/fmicb.2022.877223] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/22/2022] [Indexed: 01/15/2023] Open
Abstract
Since we previously reported that women infected with chlamydia had a significant overall reduction in Lactobacillus in the vagina microbiota as compared to those uninfected individuals; the interactions between the altered Lactobacillus and Chlamydia trachomatis, on the other hand, need to be elucidated. Here, we employed both in vitro and in vivo models to evaluate the effects of this changed Lactobacillus on Chlamydia infection. We found that L. iners, L. crispatus, L. jensenii, L. salivarius, L. gasseri, L. mucosae, and L. reuteri all significantly reduced C. trachomatis infection in a dose- and time-dependent manner. The strongest anti-Chlamydia effects were found in L. crispatus (90 percent reduction), whereas the poorest was found in L. iners (50 percent reduction). D (–) lactic acid was the key component in Lactobacillus cell-free supernatants (CFS) to inactivate Chlamydia EBs, showing a positive correlation with the anti-Chlamydia activity. The effects of D (–) lactic acid were substantially attenuated by neutralizing the pH value to 7.0. In vivo, mice intravaginally inoculated with Lactobacillus mixtures (L. crispatus, L. reuteri, and L. iners at a ratio of 1:1:1), but not single Lactobacillus, after genital Chlamydia infection, significantly attenuated the levels of Chlamydia live organism shedding in both the lower genital tract and the intestinal tract, reduced cytokines production (TNF-α, IFN-γ, and IL-1β) in the vagina, and lessened upper genital tract inflammation and pathogenicity. Taken together, these data demonstrate that Lactobacillus inhibits Chlamydia infectivity both in vivo and in vitro, providing useful information for the development of Lactobacillus as adjunctive treatment in Chlamydia infection.
Collapse
Affiliation(s)
- Hongliang Chen
- Chenzhou No.1 People's Hospital, Hengyang Medical School, University of South China, Chenzhou, China.,Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China.,Chenzhou No.1 People's Hospital, The First School of Clinical Medicine, Southern Medical University, Chenzhou, China
| | - Shuling Min
- Chenzhou No.1 People's Hospital, Hengyang Medical School, University of South China, Chenzhou, China.,Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Li Wang
- Chenzhou No.1 People's Hospital, The First School of Clinical Medicine, Southern Medical University, Chenzhou, China
| | - Lanhua Zhao
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Fangzhen Luo
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Wenbo Lei
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Yating Wen
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| | - Lipei Luo
- Chenzhou No.1 People's Hospital, The First School of Clinical Medicine, Southern Medical University, Chenzhou, China
| | - Qianting Zhou
- Chenzhou No.1 People's Hospital, Hengyang Medical School, University of South China, Chenzhou, China
| | - Lixiu Peng
- Chenzhou No.1 People's Hospital, Hengyang Medical School, University of South China, Chenzhou, China
| | - Zhongyu Li
- Institute of Pathogenic Biology, Hengyang Medical College, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, China
| |
Collapse
|
21
|
Morales B, Spadetto L, Calvo MÀ, Yeste M, Arosemena L, Rigau T, Rivera del Alamo MM. Evaluation of the Probiotic In Vitro Potential of Lactic Acid-Producing Bacteria from Canine Vagina: Possible Role in Vaginal Health. Animals (Basel) 2022; 12:ani12060796. [PMID: 35327193 PMCID: PMC8944649 DOI: 10.3390/ani12060796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/17/2022] [Accepted: 03/19/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary The use of microorganisms with probiotic capacity, obtained from the natural microbiota itself, is an alternative tool for the treatment and prevention of some animal pathologies. In this study, a total of 94 bitches were sampled in order to identify the lactic acid-producing microbiota of the vagina. Following the general isolation of the vaginal microbiota, 100 strains of lactic acid-producing bacteria (LAB) were identified. Through Gram staining and basic biochemical tests (catalase, oxidase and haemolysis), 13 LAB strains with possible probiotic capacity were selected to undergo an evaluation of resistance to gastrointestinal conditions (pH, lysozyme, bile salts and hydrogen peroxide) and safety and efficacy in vitro (resistance to antibiotics and antimicrobial capacity by disk diffusion). After the selection, only 3 of 100 strains showed in vitro probiotic potential. Abstract Lactic acid-producing bacteria (LAB) are being widely studied due to their probiotic potential. The aim of the present study was to determine and identify the presence of LAB from canine vaginal samples, as well as to evaluate their probiotic in vitro potential. Ninety-four bitches were included in the study. Vaginal samples were obtained by means of a sterile swab and streaked on Man Rogosa Sharpe agar plates. A total of 100 LAB strains were obtained and submitted to Gram stains and basic biochemical tests, which included catalase, oxidase and haemolysis tests. Thirteen strains belonging to the genera Lactobacillus (n = 10), Lactococcus (n = 2) and Pediococcus (n = 1) were selected as potential probiotics and further subjected to evaluation of resistance to gastrointestinal conditions (pH, lysozyme, bile salts and hydrogen peroxide) and safety and efficacy in vitro (resistance to antibiotics and antimicrobial capacity). Only three strains, one Lactobacillus lactis and two Lactobacillus plantarum, accomplished the requirements for being considered as potential in vitro probiotics.
Collapse
Affiliation(s)
- Brian Morales
- Department of Animal Health and Anatomy, Faculty of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (B.M.); (M.À.C.); (L.A.)
| | - Livia Spadetto
- Toxicology Research Group, Faculty of Veterinary Medicine, University of Murcia, Campus Espinardo, 30100 Murcia, Spain;
| | - Maria Àngels Calvo
- Department of Animal Health and Anatomy, Faculty of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (B.M.); (M.À.C.); (L.A.)
| | - Marc Yeste
- Department of Biology, Faculty of Sciences, University of Griona, 17071 Girona, Spain;
| | - Leonardo Arosemena
- Department of Animal Health and Anatomy, Faculty of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain; (B.M.); (M.À.C.); (L.A.)
| | - Teresa Rigau
- Department of Animal Medicine and Surgery, Faculty of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain;
- Fundació Hospital Clínic Veterinari, Faculty of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Maria Montserrat Rivera del Alamo
- Department of Animal Medicine and Surgery, Faculty of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain;
- Fundació Hospital Clínic Veterinari, Faculty of Veterinary Medicine, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Correspondence:
| |
Collapse
|
22
|
Reiprich A, Skalden L, Raab A, Bolotina N, Lang C. Lactobacillus crispatus DSM25988 as novel bioactive agent to co-aggregate Streptococcus pyogenes and to exclude it by binding to human cells. Benef Microbes 2022; 13:83-94. [PMID: 35144524 DOI: 10.3920/bm2021.0103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Streptococcus pyogenes, a group A streptococcus, is the major bacterial pathogen responsible for acute bacterial infection of the human oropharynx and the causative agent of scarlet fever. Estimates of the global burden of S. pyogenes related diseases revealed 616 million cases of pharyngitis, and at least 517,000 deaths due to severe invasive diseases and sequelae. Here we describe Lactobacillus crispatus DSM25988 that was identified among hundreds of Lactobacillus strains (referring to all organisms that were classified as Lactobacillaceae until 2020) showing ability to prevent adhesion of S. pyogenes to Detroit 562 cells, and to exhibit a masking and co-aggregating effect on S. pyogenes in vitro. L. crispatus DSM25988 also inhibits invasion of cultured human epithelial pharyngeal cells by S. pyogenes. Competitive binding to fibronectin might be involved in the inhibition process. Antiviral activity of the L. crispatus DSM25988 cells were identified in an in vitro cell model demonstrating that L. crispatus effectively excludes viruses from epithelial cells using SARS-CoV2 proteins as a model. This finding points to the potential of DSM25988 to protect cells from virus infection. Biological activity is retained in heat treated cells. The heat-treated Lactobacillus strain was further developed into functional throat lozenges, wherein its biological activity is stably maintained in the formulation. Lozenges containing L. crispatus DSM25988 underwent testing in an uncontrolled, prospective user study in 44 subjects with symptoms of sore throat for a period of up to 14 days. The study data shows promising safety and efficacy of the medical device when used against symptoms of sore throat like scratchy feeling, hoarse voice and swallowing pain.
Collapse
Affiliation(s)
- A Reiprich
- Belano medical AG, Neuendorfstraße 19, 16761 Hennigsdorf, Germany
| | - L Skalden
- Belano medical AG, Neuendorfstraße 19, 16761 Hennigsdorf, Germany
| | - A Raab
- Belano medical AG, Neuendorfstraße 19, 16761 Hennigsdorf, Germany
| | - N Bolotina
- Belano medical AG, Neuendorfstraße 19, 16761 Hennigsdorf, Germany
| | - C Lang
- Belano medical AG, Neuendorfstraße 19, 16761 Hennigsdorf, Germany
| |
Collapse
|
23
|
Unravelling the functional and technological potential of soy milk based microencapsulated Lactobacillus crispatus and Lactobacillus gasseri. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104745] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
24
|
Ansari A, Bose S, You Y, Park S, Kim Y. Molecular Mechanism of Microbiota Metabolites in Preterm Birth: Pathological and Therapeutic Insights. Int J Mol Sci 2021; 22:8145. [PMID: 34360908 PMCID: PMC8347546 DOI: 10.3390/ijms22158145] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 12/11/2022] Open
Abstract
Preterm birth (PTB) refers to the birth of infants before 37 weeks of gestation and is a challenging issue worldwide. Evidence reveals that PTB is a multifactorial dysregulation mediated by a complex molecular mechanism. Thus, a better understanding of the complex molecular mechanisms underlying PTB is a prerequisite to explore effective therapeutic approaches. During early pregnancy, various physiological and metabolic changes occur as a result of endocrine and immune metabolism. The microbiota controls the physiological and metabolic mechanism of the host homeostasis, and dysbiosis of maternal microbial homeostasis dysregulates the mechanistic of fetal developmental processes and directly affects the birth outcome. Accumulating evidence indicates that metabolic dysregulation in the maternal or fetal membranes stimulates the inflammatory cytokines, which may positively progress the PTB. Although labour is regarded as an inflammatory process, it is still unclear how microbial dysbiosis could regulate the molecular mechanism of PTB. In this review based on recent research, we focused on both the pathological and therapeutic contribution of microbiota-generated metabolites to PTB and the possible molecular mechanisms.
Collapse
Affiliation(s)
- AbuZar Ansari
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Mokdong Hospital, Seoul 07985, Korea; (A.A.); (Y.Y.); (S.P.)
| | - Shambhunath Bose
- Department of Bioscience, Sri Sathya Sai University for Human Excellence, Navanihal, Okali Post, Kamalapur, Kalaburagi, Karnataka 585313, India;
| | - Youngah You
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Mokdong Hospital, Seoul 07985, Korea; (A.A.); (Y.Y.); (S.P.)
| | - Sunwha Park
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Mokdong Hospital, Seoul 07985, Korea; (A.A.); (Y.Y.); (S.P.)
| | - Youngju Kim
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, College of Medicine, Ewha Womans University, Mokdong Hospital, Seoul 07985, Korea; (A.A.); (Y.Y.); (S.P.)
| |
Collapse
|
25
|
Costantini PE, Firrincieli A, Fedi S, Parolin C, Viti C, Cappelletti M, Vitali B. Insight into phenotypic and genotypic differences between vaginal Lactobacillus crispatus BC5 and Lactobacillus gasseri BC12 to unravel nutritional and stress factors influencing their metabolic activity. Microb Genom 2021; 7. [PMID: 34096840 PMCID: PMC8461478 DOI: 10.1099/mgen.0.000575] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
The vaginal microbiota, normally characterized by lactobacilli presence, is crucial for vaginal health. Members belonging to L. crispatus and L. gasseri species exert crucial protective functions against pathogens, although a total comprehension of factors that influence their dominance in healthy women is still lacking. Here we investigated the complete genome sequence and comprehensive phenotypic profile of L. crispatus strain BC5 and L. gasseri strain BC12, two vaginal strains featured by anti-bacterial and anti-viral activities. Phenotype microarray (PM) results revealed an improved capacity of BC5 to utilize different carbon sources as compared to BC12, although some specific carbon sources that can be associated to the human diet were only metabolized by BC12, i.e. uridine, amygdalin, tagatose. Additionally, the two strains were mostly distinct in the capacity to utilize the nitrogen sources under analysis. On the other hand, BC12 showed tolerance/resistance towards twice the number of stressors (i.e. antibiotics, toxic metals etc.) with respect to BC5. The divergent phenotypes observed in PM were supported by the identification in either BC5 or BC12 of specific genetic determinants that were found to be part of the core genome of each species. The PM results in combination with comparative genome data provide insights into the possible environmental factors and genetic traits supporting the predominance of either L. crispatus BC5 or L. gasseri BC12 in the vaginal niche, giving also indications for metabolic predictions at the species level.
Collapse
Affiliation(s)
| | - Andrea Firrincieli
- Department of Pharmacy and Biotechnology (FaBit), University of Bologna, Bologna, Italy
| | - Stefano Fedi
- Department of Pharmacy and Biotechnology (FaBit), University of Bologna, Bologna, Italy
| | - Carola Parolin
- Department of Pharmacy and Biotechnology (FaBit), University of Bologna, Bologna, Italy
| | - Carlo Viti
- Department of Agriculture, Food, Environment and Forestry (DAGRI), University of Florence, Florence, Italy
| | - Martina Cappelletti
- Department of Pharmacy and Biotechnology (FaBit), University of Bologna, Bologna, Italy
| | - Beatrice Vitali
- Department of Pharmacy and Biotechnology (FaBit), University of Bologna, Bologna, Italy
| |
Collapse
|
26
|
Foschi C, Parolin C, Giordani B, Morselli S, Luppi B, Vitali B, Marangoni A. Lactobacillus crispatus BC1 Biosurfactant Counteracts the Infectivity of Chlamydia trachomatis Elementary Bodies. Microorganisms 2021; 9:microorganisms9050975. [PMID: 33946391 PMCID: PMC8147163 DOI: 10.3390/microorganisms9050975] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/19/2021] [Accepted: 04/27/2021] [Indexed: 01/01/2023] Open
Abstract
Lactobacilli-derived biosurfactants (BS) have shown promising effects as antimicrobial molecules. Since Lactobacillus crispatus plays a crucial role in maintaining vaginal eubiosis, BS from this species could represent novel therapeutic agents to counteract sexually transmitted pathogens, such as Chlamydia trachomatis (CT). The aim of the present study was to assess the inhibitory effects of a BS produced by the vaginal strain L. crispatus BC1 on the infectivity of CT elementary bodies (EBs). For concentrations ranging between 1 and 0.5 mg/mL at 60-min contact time, L. crispatus BC1 BS displayed a highly significant anti-CT activity, with about 50% reduction of EB infectivity towards HeLa cells. To identify the components responsible for chlamydial inhibition, a panel of selected fatty acids, including those present in BS lipopeptidic structure, was tested against CT EBs. Pentadecanoic acid, myristic acid, β-hydroxy-myristic acid, and β-hydroxy-palmitic acid were able to significantly reduce EBs infectivity up to 5–0.5 µg/mL, concentrations that resulted to be non-toxic for HeLa cells. These data can contribute to the understanding of the biological role of lactobacilli in the vaginal niche, as well as to promote the application of their produced BS as an innovative and antibiotic-sparing anti-chlamydial strategy.
Collapse
Affiliation(s)
- Claudio Foschi
- Microbiology, DIMES, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (C.F.); (S.M.); (A.M.)
| | - Carola Parolin
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, 40127 Bologna, Italy; (B.G.); (B.L.); (B.V.)
- Correspondence: ; Tel.: +39-051-2088750
| | - Barbara Giordani
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, 40127 Bologna, Italy; (B.G.); (B.L.); (B.V.)
| | - Sara Morselli
- Microbiology, DIMES, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (C.F.); (S.M.); (A.M.)
| | - Barbara Luppi
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, 40127 Bologna, Italy; (B.G.); (B.L.); (B.V.)
| | - Beatrice Vitali
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum, University of Bologna, 40127 Bologna, Italy; (B.G.); (B.L.); (B.V.)
| | - Antonella Marangoni
- Microbiology, DIMES, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy; (C.F.); (S.M.); (A.M.)
| |
Collapse
|
27
|
D’Alessandro M, Parolin C, Bukvicki D, Siroli L, Vitali B, De Angelis M, Lanciotti R, Patrignani F. Probiotic and Metabolic Characterization of Vaginal Lactobacilli for a Potential Use in Functional Foods. Microorganisms 2021; 9:microorganisms9040833. [PMID: 33919838 PMCID: PMC8070825 DOI: 10.3390/microorganisms9040833] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 04/12/2021] [Indexed: 12/04/2022] Open
Abstract
The main aim of this work was to verify the metabolic and functional aptitude of 15 vaginal strains belonging to Lactobacillus crispatus, Lactobacillus gasseri, and Limosilactobacillus vaginalis (previously Lactobacillus vaginalis), already characterized for their technological and antimicrobial properties. In order to evaluate the metabolic profile of these vaginal strains, a phenotype microarray analysis was performed on them. Functional parameters such as hydrophobicity, auto-aggregation, deconjugation of bile salts, adhesion to an intestinal cell line (Caco-2), and a simulated digestion process were evaluated for these strains. A good number of these strains showed hydrophobicity values higher than 70%. Regarding the auto-aggregation assay, the most promising strains were L. crispatus BC9 and BC1, L. gasseri BC10 and BC14, and L. vaginalis BC17. Moreover, L. crispatus BC4, BC6, BC7, and BC8 were characterized by strong bile salts hydrolase activity (BHS). In addition, L. crispatus BC8 and L. vaginalis BC17 were characterized by a medium ability to adhere to Caco-2 cells. Data related to digestion process showed a strong ability of vaginal lactobacilli to withstand this stress. In conclusion, the data collected show the metabolic versatility and several exploitable functional properties of the investigated vaginal lactobacilli.
Collapse
Affiliation(s)
- Margherita D’Alessandro
- Department of Agricultural and Food Sciences, University of Bologna, 47521 Cesena, Italy; (L.S.); (R.L.); (F.P.)
- Correspondence:
| | - Carola Parolin
- Department of Pharmacy and Biotechnology, University of Bologna, 40127 Bologna, Italy; (C.P.); (B.V.)
| | - Danka Bukvicki
- Faculty of Biology, Institute of Botany and Botanical Garden “Jevremovac”, University of Belgrade, 11000 Belgrade, Serbia;
| | - Lorenzo Siroli
- Department of Agricultural and Food Sciences, University of Bologna, 47521 Cesena, Italy; (L.S.); (R.L.); (F.P.)
- Interdepartmental Center for Industrial Agri-Food Research, University of Bologna, 47521 Cesena, Italy
| | - Beatrice Vitali
- Department of Pharmacy and Biotechnology, University of Bologna, 40127 Bologna, Italy; (C.P.); (B.V.)
| | - Maria De Angelis
- Department of Soil, Plant, and Food Sciences, University of Bari Aldo Moro, 70126 Bari, Italy;
| | - Rosalba Lanciotti
- Department of Agricultural and Food Sciences, University of Bologna, 47521 Cesena, Italy; (L.S.); (R.L.); (F.P.)
- Interdepartmental Center for Industrial Agri-Food Research, University of Bologna, 47521 Cesena, Italy
| | - Francesca Patrignani
- Department of Agricultural and Food Sciences, University of Bologna, 47521 Cesena, Italy; (L.S.); (R.L.); (F.P.)
- Interdepartmental Center for Industrial Agri-Food Research, University of Bologna, 47521 Cesena, Italy
| |
Collapse
|
28
|
Lactobacillus crispatus BC1 Biosurfactant Delivered by Hyalurosomes: An Advanced Strategy to Counteract Candida Biofilm. Antibiotics (Basel) 2021; 10:antibiotics10010033. [PMID: 33401413 PMCID: PMC7823809 DOI: 10.3390/antibiotics10010033] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 12/28/2020] [Accepted: 12/29/2020] [Indexed: 02/06/2023] Open
Abstract
The emergence of resistance to antifungal drugs has made the treatment of vulvovaginal candidiasis (VVC) very challenging. Among natural substances, biosurfactants (BS) produced by Lactobacillus have gained increasing interest in counteracting Candida infections for their proven anti-adhesive properties and safety profile. In the present study, liposomes (LP-BS) or liposomes coated with hyaluronic acid (HY-LP-BS) were prepared in the presence of the BS isolated from the vaginal strain Lactobacillus crispatus BC1 and characterized in terms of size, ζ potential, stability and mucoadhesion. The anti-biofilm activity of free BS, LP-BS and HY-LP-BS was investigated against different Candida albicans and non-albicans strains (C. glabrata, C. lusitaniae, C. tropicalis, C. krusei and C. parapsilosis), clinically isolated from patients affected by VVC. The inhibition of biofilm formation and the dispersal of pre-formed biofilm were evaluated. The obtained phospholipid vesicles showed suitable size for vaginal application and good stability over the storage period. HY-LP-BS exhibited good mucoadhesive properties and the best anti-biofilm profile, both in preventing or limiting the surface colonization by a broad spectrum of Candida species. In conclusion, the formulation of a novel antifungal agent derived from the vaginal microbiota into mucoadhesive nanocarriers appears to be a promising biotherapeutic strategy to counteract vulvovaginal candidiasis.
Collapse
|
29
|
Yuan L, van der Mei HC, Busscher HJ, Peterson BW. Two-Stage Interpretation of Changes in TEER of Intestinal Epithelial Layers Protected by Adhering Bifidobacteria During E. coli Challenges. Front Microbiol 2020; 11:599555. [PMID: 33329490 PMCID: PMC7710611 DOI: 10.3389/fmicb.2020.599555] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 10/26/2020] [Indexed: 12/19/2022] Open
Abstract
Mechanisms of gastrointestinal protection by probiotic bacteria against infection involve amongst others, modulation of intestinal epithelial barrier function. Trans-epithelial electrical resistance (TEER) is widely used to evaluate cellular barrier functions. Here, we developed a two-stage interpretative model of the time-dependence of the TEER of epithelial layers grown in a transwell during Escherichia coli challenges in the absence or presence of adhering bifidobacteria. E. coli adhesion in absence or presence of adhering bifidobacteria was enumerated using selective plating. After 4-8 h, E. coli challenges increased TEER to a maximum due to bacterial adhesion and increased expression of a tight-junction protein [zonula occludens-1 (ZO-1)], concurrent with a less dense layer structure, that is indicative of mild epithelial layer damage. Before the occurrence of a TEER-maximum, decreases in electrical conductance (i.e., the reciprocal TEER) did not relate with para-cellular dextran-permeability, but after occurrence of a TEER-maximum, dextran-permeability and conductance increased linearly, indicative of more severe epithelial layer damage. Within 24 h after the occurrence of a TEER maximum, TEER decreased to below the level of unchallenged epithelial layers demonstrating microscopically observable holes and apoptosis. Under probiotic protection by adhering bifidobacteria, TEER-maxima were delayed or decreased in magnitude due to later transition from mild to severe damage, but similar linear relations between conductance and dextran permeability were observed as in absence of adhering bifidobacteria. Based on the time-dependence of the TEER and the relation between conductance and dextran-permeability, it is proposed that bacterial adhesion to epithelial layers first causes mild damage, followed by more severe damage after the occurrence of a TEER-maximum. The mild damage caused by E. coli prior to the occurrence of TEER maxima was reversible upon antibiotic treatment, but the severe damage after occurrence of TEER maxima could not be reverted by antibiotic treatment. Thus, single-time TEER is interpretable in two ways, depending whether increasing to or decreasing from its maximum. Adhering bifidobacteria elongate the time-window available for antibiotic treatment to repair initial pathogen damage to intestinal epithelial layers.
Collapse
Affiliation(s)
| | | | | | - Brandon W. Peterson
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
30
|
De Marco R, Rampazzo E, Zhao J, Prodi L, Paolillo M, Picchetti P, Gallo F, Calonghi N, Gentilucci L. Integrin-Targeting Dye-Doped PEG-Shell/Silica-Core Nanoparticles Mimicking the Proapoptotic Smac/DIABLO Protein. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1211. [PMID: 32575872 PMCID: PMC7353088 DOI: 10.3390/nano10061211] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/14/2020] [Accepted: 06/18/2020] [Indexed: 01/08/2023]
Abstract
Cancer cells demonstrate elevated expression levels of the inhibitor of apoptosis proteins (IAPs), contributing to tumor cell survival, disease progression, chemo-resistance, and poor prognosis. Smac/DIABLO is a mitochondrial protein that promotes apoptosis by neutralizing members of the IAP family. Herein, we describe the preparation and in vitro validation of a synthetic mimic of Smac/DIABLO, based on fluorescent polyethylene glycol (PEG)-coated silica-core nanoparticles (NPs) carrying a Smac/DIABLO-derived pro-apoptotic peptide and a tumor-homing integrin peptide ligand. At low μM concentration, the NPs showed significant toxicity towards A549, U373, and HeLa cancer cells and modest toxicity towards other integrin-expressing cells, correlated with integrin-mediated cell uptake and consequent highly increased levels of apoptotic activity, without perturbing cells not expressing the α5 integrin subunit.
Collapse
Affiliation(s)
- Rossella De Marco
- Department of Agricultural, Food, Enviromental and Animal Sciences (DI4A), University of Udine, 33100 Udine, Italy;
| | - Enrico Rampazzo
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (E.R.); (J.Z.); (L.P.); (F.G.)
| | - Junwei Zhao
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (E.R.); (J.Z.); (L.P.); (F.G.)
| | - Luca Prodi
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (E.R.); (J.Z.); (L.P.); (F.G.)
| | - Mayra Paolillo
- Department of Drugs Sciences, University of Pavia, 27100 Pavia, Italy;
| | - Pierre Picchetti
- Institut de Science et d’Ingénierie Supramoléculaires (ISIS), Université de Strasbourg, 67083 Strasbourg, France;
| | - Francesca Gallo
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (E.R.); (J.Z.); (L.P.); (F.G.)
| | - Natalia Calonghi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Luca Gentilucci
- Department of Chemistry “G. Ciamician”, University of Bologna, 40126 Bologna, Italy; (E.R.); (J.Z.); (L.P.); (F.G.)
| |
Collapse
|
31
|
De Gregorio PR, Parolin C, Abruzzo A, Luppi B, Protti M, Mercolini L, Silva JA, Giordani B, Marangoni A, Nader-Macías MEF, Vitali B. Biosurfactant from vaginal Lactobacillus crispatus BC1 as a promising agent to interfere with Candida adhesion. Microb Cell Fact 2020; 19:133. [PMID: 32552788 PMCID: PMC7302142 DOI: 10.1186/s12934-020-01390-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 06/11/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Lactobacillus spp. dominating the vaginal microbiota of healthy women contribute to the prevention of urogenital and sexually transmitted infections. Their protective role in the vagina can be mediated by Lactobacillus cells themselves, metabolites or bacterial components, able to interfere with pathogen adhesion and infectivity. Vulvovaginal candidiasis (VVC) is a common genital infection, caused by the overgrowth of opportunistic Candida spp. including C. albicans, C. glabrata, C. krusei and C. tropicalis. Azole antifungal drugs are not always efficient in resolving VVC and preventing recurrent infections, thus alternative anti-Candida agents based on vaginal probiotics have gained more importance. The present work aims to chemically characterize the biosurfactant (BS) isolated from a vaginal Lactobacillus crispatus strain, L. crispatus BC1, and to investigate its safety and antiadhesive/antimicrobial activity against Candida spp., employing in vitro and in vivo assays. RESULTS BS isolated from vaginal L. crispatus BC1 was characterised as non-homogeneous lipopeptide molecules with a critical micellar concentration value of 2 mg/mL, and good emulsification and mucoadhesive properties. At 1.25 mg/mL, the BS was not cytotoxic and reduced Candida strains' ability to adhere to human cervical epithelial cells, mainly by exclusion mechanism. Moreover, intravaginal (i.va.) inoculation of BS in a murine experimental model was safe and did not perturb vaginal cytology, histology and cultivable vaginal microbiota. In the case of i.va. challenge of mice with C. albicans, BS was able to reduce leukocyte influx. CONCLUSIONS These results indicate that BS from vaginal L. crispatus BC1 is able to interfere with Candida adhesion in vitro and in vivo, and suggest its potential as a preventive agent to reduce mucosal damage occasioned by Candida during VVC.
Collapse
Affiliation(s)
- Priscilla Romina De Gregorio
- Centro de Referencia para Lactobacilos (CERELA)-CONICET, Chacabuco, 145, 4000, San Miguel de Tucumán, Tucumán, Argentina.
| | - Carola Parolin
- Department of Pharmacy and Biotechnologies, University of Bologna, Via San Donato 19/2, 40127, Bologna, Italy.
| | - Angela Abruzzo
- Department of Pharmacy and Biotechnologies, University of Bologna, Via San Donato 19/2, 40127, Bologna, Italy
| | - Barbara Luppi
- Department of Pharmacy and Biotechnologies, University of Bologna, Via San Donato 19/2, 40127, Bologna, Italy
| | - Michele Protti
- Department of Pharmacy and Biotechnologies, University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Laura Mercolini
- Department of Pharmacy and Biotechnologies, University of Bologna, Via Belmeloro 6, 40126, Bologna, Italy
| | - Jessica Alejandra Silva
- Centro de Referencia para Lactobacilos (CERELA)-CONICET, Chacabuco, 145, 4000, San Miguel de Tucumán, Tucumán, Argentina
| | - Barbara Giordani
- Department of Pharmacy and Biotechnologies, University of Bologna, Via San Donato 19/2, 40127, Bologna, Italy
| | - Antonella Marangoni
- Microbiology, DIMES, University of Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | | | - Beatrice Vitali
- Department of Pharmacy and Biotechnologies, University of Bologna, Via San Donato 19/2, 40127, Bologna, Italy
| |
Collapse
|
32
|
Marziali G, Marangoni A, Foschi C, Re MC, Calonghi N. Effect of Sugars on Chlamydia trachomatis Infectivity. Pathogens 2020; 9:pathogens9040298. [PMID: 32316668 PMCID: PMC7237991 DOI: 10.3390/pathogens9040298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/09/2020] [Accepted: 04/16/2020] [Indexed: 11/16/2022] Open
Abstract
Background. Previous works suggest that sugars can have a beneficial effect on C. trachomatis (CT) survival and virulence. In this study, we investigated the effect of different sugars on CT infectivity, elucidating some of the molecular mechanisms behind CT-sugar interaction. Methods. CT infectivity was investigated on HeLa cells after 2 hour-incubation of elementary bodies (EBs) with glucose, sucrose, or mannitol solutions (0.5, 2.5, 5.0 mM). The effect of sugars on EB membrane fluidity was investigated by fluorescence anisotropy measurement, whereas the changes in lipopolysaccharide (LPS) exposure were examined by cytofluorimetric analysis. By means of a Western blot, we explored the phosphorylation state of Focal Adhesion Kinase (FAK) in HeLa cells infected with EBs pre-incubated with sugars. Results. All sugar solutions significantly increased CT infectivity on epithelial cells, acting directly on the EB structure. Sugars induced a significant increase of EB membrane fluidity, leading to changes in LPS membrane exposure. Especially after incubation with sucrose and mannitol, EBs led to a higher FAK phosphorylation, enhancing the activation of anti-apoptotic and proliferative signals in the host cells. Conclusions. Sugars can increase CT infectivity and virulence, by modulating the expression/exposure of chlamydial membrane ligands. Further in-depth studies are needed to better understand the molecular mechanisms involved.
Collapse
Affiliation(s)
- Giacomo Marziali
- FaBiT Department, University of Bologna, Via Irnerio 42, 40126 Bologna, Italy; (G.M.); (N.C.)
| | - Antonella Marangoni
- Microbiology, DIMES, University of Bologna, St. Orsola Hospital, Via Massarenti, 9, 40138 Bologna, Italy; (A.M.); (M.C.R.)
| | - Claudio Foschi
- Microbiology, DIMES, University of Bologna, St. Orsola Hospital, Via Massarenti, 9, 40138 Bologna, Italy; (A.M.); (M.C.R.)
- Correspondence: ; Tel.: +39-051-2144513; Fax: +39-051-307397
| | - Maria Carla Re
- Microbiology, DIMES, University of Bologna, St. Orsola Hospital, Via Massarenti, 9, 40138 Bologna, Italy; (A.M.); (M.C.R.)
| | - Natalia Calonghi
- FaBiT Department, University of Bologna, Via Irnerio 42, 40126 Bologna, Italy; (G.M.); (N.C.)
| |
Collapse
|
33
|
Draft Genome Sequence of Lactobacillus crispatus CIP 104459, Isolated from a Vaginal Swab. Microbiol Resour Announc 2020; 9:9/2/e01373-19. [PMID: 31919178 PMCID: PMC6952664 DOI: 10.1128/mra.01373-19] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
We report the draft genome sequence of Lactobacillus crispatus CIP 104459, isolated from a human vaginal swab. This draft genome consists of 1,993,673 bp, with 36.8% G+C content and 2,024 predicted protein-encoding sequences. We report the draft genome sequence of Lactobacillus crispatus CIP 104459, isolated from a human vaginal swab. This draft genome consists of 1,993,673 bp, with 36.8% G+C content and 2,024 predicted protein-encoding sequences.
Collapse
|
34
|
Extracellular vesicles from symbiotic vaginal lactobacilli inhibit HIV-1 infection of human tissues. Nat Commun 2019; 10:5656. [PMID: 31827089 PMCID: PMC6906448 DOI: 10.1038/s41467-019-13468-9] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 11/07/2019] [Indexed: 12/20/2022] Open
Abstract
The vaginal microbiota, dominated by Lactobacillus spp., plays a key role in preventing HIV-1 transmission. Here, we investigate whether the anti-HIV effect of lactobacilli is mediated by extracellular vesicles (EVs) released by these bacteria. Human cervico-vaginal and tonsillar tissues ex vivo, and cell lines were infected with HIV-1 and treated with EVs released by lactobacilli isolated from vaginas of healthy women. EVs released by L. crispatus BC3 and L. gasseri BC12 protect tissues ex vivo and isolated cells from HIV-1 infection. This protection is associated with a decrease of viral attachment to target cells and viral entry due to diminished exposure of Env that mediates virus-cell interactions. Inhibition of HIV-1 infection is associated with the presence in EVs of several proteins and metabolites. Our findings demonstrate that the protective effect of Lactobacillus against HIV-1 is, in part, mediated by EVs released by these symbiotic bacteria. If confirmed in vivo, this finding may lead to new strategies to prevent male-to-female sexual HIV-1 transmission. Lactobacillus associates with vaginal protection from HIV-1 infection. Here, the authors show that lactobacilli extracellular vesicles contain bacterial proteins and metabolites that inhibit HIV-1 infection in T cells and in human cervico-vaginal and tonsillar tissues ex vivo via altering viral Env proteins.
Collapse
|
35
|
De Seta F, Campisciano G, Zanotta N, Ricci G, Comar M. The Vaginal Community State Types Microbiome-Immune Network as Key Factor for Bacterial Vaginosis and Aerobic Vaginitis. Front Microbiol 2019; 10:2451. [PMID: 31736898 PMCID: PMC6831638 DOI: 10.3389/fmicb.2019.02451] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 10/11/2019] [Indexed: 01/19/2023] Open
Abstract
Regarding bacterial vaginosis (BV), the relevance of the vaginal microbiota to the women’s health fulfills a key role, but knowledge gaps regarding aerobic vaginitis (AV) exist. This study aims to characterize vaginal microbiome and its relationship with the local immune mediators, providing an opportunity to define the link between vaginal commensal microorganisms and opportunistic pathogens in the relation of a given vaginal community state type (CST). A total of 90 vaginal samples from Caucasian asymptomatic women of reproductive age (18–40 years) attending the yearly examination and not reporting any vaginal complaints were retrospectively evaluated for microbiome assessment and immune factor dosage. The samples were tested by the Ion Torrent PGM and the Luminex Bio-Plex technologies for the analysis of microbiome and immune factors, respectively. In our study, the CST classification together with the local immune response profiling represented a good predictive indicator of the vaginal health, suggesting that the predominance of a specific Lactobacillus and its relative abundance are pivotal elements to maintain a physiologic status. A vaginal colonization from Bifidobacterium may absolve a protective role similar to that of Lactobacillus, corresponding to a newly identified CST, although studies are needed to better clarify its clinical significance. Moreover, within each CST, a different pattern of inflammation is activated and orchestrated both by the dominant Lactobacillus spp. and by specific non-Lactobacillus bacteria and can give insights into the pathogenic mechanisms. In conclusion, this study contributes to the characterization of vaginal dysbiosis, reshaping this concept by taking into consideration the CST profiling, local immune marker, and immune–microbial network.
Collapse
Affiliation(s)
- Francesco De Seta
- Institute for Maternal and Child Health "IRCCS Burlo Garofolo", Trieste, Italy.,Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | | | - Nunzia Zanotta
- Institute for Maternal and Child Health "IRCCS Burlo Garofolo", Trieste, Italy
| | - Giuseppe Ricci
- Institute for Maternal and Child Health "IRCCS Burlo Garofolo", Trieste, Italy.,Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| | - Manola Comar
- Institute for Maternal and Child Health "IRCCS Burlo Garofolo", Trieste, Italy.,Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy
| |
Collapse
|
36
|
Ceccarani C, Foschi C, Parolin C, D'Antuono A, Gaspari V, Consolandi C, Laghi L, Camboni T, Vitali B, Severgnini M, Marangoni A. Diversity of vaginal microbiome and metabolome during genital infections. Sci Rep 2019; 9:14095. [PMID: 31575935 PMCID: PMC6773718 DOI: 10.1038/s41598-019-50410-x] [Citation(s) in RCA: 183] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 09/06/2019] [Indexed: 12/30/2022] Open
Abstract
We characterized the vaginal ecosystem during common infections of the female genital tract, as vulvovaginal candidiasis (VVC, n = 18) and Chlamydia trachomatis infection (CT, n = 20), recruiting healthy (HC, n = 21) and bacterial vaginosis-affected (BV, n = 20) women as references of eubiosis and dysbiosis. The profiles of the vaginal microbiome and metabolome were studied in 79 reproductive-aged women, by means of next generation sequencing and proton based-nuclear magnetic resonance spectroscopy. Lactobacillus genus was profoundly depleted in all the genital infections herein considered, and species-level analysis revealed that healthy vaginal microbiome was dominated by L. crispatus. In the shift from HC to CT, VVC, and BV, L. crispatus was progressively replaced by L. iners. CT infection and VVC, as well as BV condition, were mainly characterised by anaerobe genera, e.g. Gardnerella, Prevotella, Megasphaera, Roseburia and Atopobium. The changes in the bacterial communities occurring during the genital infections resulted in significant alterations in the vaginal metabolites composition, being the decrease of lactate a common marker of all the pathological conditions. In conclusion, according to the taxonomic and metabolomics analysis, we found that each of the four conditions is characterized by a peculiar vaginal microbiome/metabolome fingerprint.
Collapse
Affiliation(s)
- Camilla Ceccarani
- Institute of Biomedical Technologies, National Research Council, Segrate, Milan, Italy.,Department of Health Sciences, San Paolo Hospital Medical School, University of Milan, Milan, Italy
| | - Claudio Foschi
- Microbiology, Experimental Diagnostic and Specialty Department (DIMES), University of Bologna, Bologna, Italy
| | - Carola Parolin
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy.
| | | | | | - Clarissa Consolandi
- Institute of Biomedical Technologies, National Research Council, Segrate, Milan, Italy
| | - Luca Laghi
- Centre of Foodomics, Department of Agricultural and Food Sciences, University of Bologna, Cesena, Italy
| | - Tania Camboni
- Institute of Biomedical Technologies, National Research Council, Segrate, Milan, Italy
| | - Beatrice Vitali
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Bologna, Italy
| | - Marco Severgnini
- Institute of Biomedical Technologies, National Research Council, Segrate, Milan, Italy
| | - Antonella Marangoni
- Microbiology, Experimental Diagnostic and Specialty Department (DIMES), University of Bologna, Bologna, Italy
| |
Collapse
|
37
|
Pekmezovic M, Mogavero S, Naglik JR, Hube B. Host-Pathogen Interactions during Female Genital Tract Infections. Trends Microbiol 2019; 27:982-996. [PMID: 31451347 DOI: 10.1016/j.tim.2019.07.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 06/25/2019] [Accepted: 07/22/2019] [Indexed: 12/11/2022]
Abstract
Dysbiosis in the female genital tract (FGT) is characterized by the overgrowth of pathogenic bacterial, fungal, or protozoan members of the microbiota, leading to symptomatic or asymptomatic infections. In this review, we discuss recent advances in studies dealing with molecular mechanisms of pathogenicity factors of Gardnerella vaginalis, Mycoplasma genitalium, Mycoplasma hominis, Neisseria gonorrhoeae, Streptococcus agalactiae, Chlamydia trachomatis, Trichomonas vaginalis, and Candida spp., as well as their interactions with the host and microbiota in the various niches of the FGT. Taking a holistic approach to identifying fundamental commonalities and differences during these infections could help us to better understand reproductive tract health and improve current prevention and treatment strategies.
Collapse
Affiliation(s)
- Marina Pekmezovic
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Selene Mogavero
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dental, Oral, and Craniofacial Sciences, King's College London, SE1 1UL, UK
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Hans Knoell Institute, Jena, Germany; Institute of Microbiology, Friedrich Schiller University, Jena, Germany. @leibniz-hki.de
| |
Collapse
|
38
|
Marziali G, Foschi C, Parolin C, Vitali B, Marangoni A. In-vitro effect of vaginal lactobacilli against group B Streptococcus. Microb Pathog 2019; 136:103692. [PMID: 31445119 DOI: 10.1016/j.micpath.2019.103692] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/29/2019] [Accepted: 08/21/2019] [Indexed: 01/31/2023]
Abstract
Streptococcus agalactiae(GBS) is a leading cause of infection during pregnancy, preterm birth and neonatal infection, with a significant clinical and socio-economic impact. To prevent maternal GBS vaginal colonization, new antibiotic-free approaches, based on lactobacilli probiotics, are advisable. The aim of this study was to assess the anti-GBS activity of 14 vaginal Lactobacillus strains, belonging to different species (L. crispatus, L. gasseri, L. vaginalis), isolated from healthy pre-menopausal women. In particular, we performed 'inhibition' experiments, evaluating the ability of both Lactobacillus cells and culture supernatants in reducing Streptococcus viability, after 60 min contact time. First, we demonstrated that the acidic milieu, produced by vaginal lactobacilli metabolism, is crucial in counteracting GBS growth in a pH-dependent manner. Experiments with organic/inorganic acid solutions confirmed the strict correlation between pH levels and the anti-GBS activity. GBS was more sensitive to lactic acid than to hydrochloric acid, indicating that the presence of H+ ions is necessary but not sufficient for the inhibitory activity. Moreover, experiments with Lactobacillus pH-adjusted supernatants led to exclude a direct role in the anti-GBS activity by other bioactive molecules. Second, we found that only a few Lactobacillus strains were able to reduce Streptococcus viability by means of cell pellets. The anti-GBS effect displayed by Lactobacillus cells was related to the their ability to interact and aggregate with Streptococcus cells. We found that the anti-GBS activity was retained after methanol/proteinase K treatment, but lost after lysozyme exposure of Lactobacillus cells. Therefore, we supposed that non-proteinaceous components of Lactobacillus cell wall could be responsible for the anti-GBS activity. In conclusion, we identified specific Lactobacillus strains able to interfere with GBS viability by multiple strategies and we elucidated some of the mechanisms of action. These strains could serve as probiotic formulations for the prevention of GBS vaginal colonization.
Collapse
Affiliation(s)
- Giacomo Marziali
- Microbiology, DIMES, University of Bologna, Via Massarenti 9, Bologna, Italy
| | - Claudio Foschi
- Microbiology, DIMES, University of Bologna, Via Massarenti 9, Bologna, Italy.
| | - Carola Parolin
- Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 19/2, Bologna, Italy
| | - Beatrice Vitali
- Department of Pharmacy and Biotechnology, University of Bologna, Via San Donato 19/2, Bologna, Italy
| | - Antonella Marangoni
- Microbiology, DIMES, University of Bologna, Via Massarenti 9, Bologna, Italy
| |
Collapse
|
39
|
Hidalgo-Cantabrana C, Goh YJ, Pan M, Sanozky-Dawes R, Barrangou R. Genome editing using the endogenous type I CRISPR-Cas system in Lactobacillus crispatus. Proc Natl Acad Sci U S A 2019; 116:15774-15783. [PMID: 31341082 PMCID: PMC6690032 DOI: 10.1073/pnas.1905421116] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
CRISPR-Cas systems are now widely used for genome editing and transcriptional regulation in diverse organisms. The compact and portable nature of class 2 single effector nucleases, such as Cas9 or Cas12, has facilitated directed genome modifications in plants, animals, and microbes. However, most CRISPR-Cas systems belong to the more prevalent class 1 category, which hinges on multiprotein effector complexes. In the present study, we detail how the native type I-E CRISPR-Cas system, with a 5'-AAA-3' protospacer adjacent motif (PAM) and a 61-nucleotide guide CRISPR RNA (crRNA) can be repurposed for efficient chromosomal targeting and genome editing in Lactobacillus crispatus, an important commensal and beneficial microbe in the vaginal and intestinal tracts. Specifically, we generated diverse mutations encompassing a 643-base pair (bp) deletion (100% efficiency), a stop codon insertion (36%), and a single nucleotide substitution (19%) in the exopolysaccharide priming-glycosyl transferase (p-gtf). Additional genetic targets included a 308-bp deletion (20%) in the prophage DNA packaging Nu1 and a 730-bp insertion of the green fluorescent protein gene downstream of enolase (23%). This approach enables flexible alteration of the formerly genetically recalcitrant species L. crispatus, with potential for probiotic enhancement, biotherapeutic engineering, and mucosal vaccine delivery. These results also provide a framework for repurposing endogenous CRISPR-Cas systems for flexible genome targeting and editing, while expanding the toolbox to include one of the most abundant and diverse systems found in nature.
Collapse
Affiliation(s)
- Claudio Hidalgo-Cantabrana
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC 27695
| | - Yong Jun Goh
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC 27695
| | - Meichen Pan
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC 27695
| | - Rosemary Sanozky-Dawes
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC 27695
| | - Rodolphe Barrangou
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC 27695
| |
Collapse
|
40
|
Verma R, Sahu R, Singh DD, Egbo TE. A CRISPR/Cas9 based polymeric nanoparticles to treat/inhibit microbial infections. Semin Cell Dev Biol 2019; 96:44-52. [PMID: 30986568 DOI: 10.1016/j.semcdb.2019.04.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/11/2019] [Indexed: 12/17/2022]
Abstract
The latest breakthrough towards the adequate and decisive methods of gene editing tools provided by CRISPR/Cas9 (Clustered Regularly Interspaced Short Palindromic Repeat/CRISPR Associated System), has been repurposed into a tool for genetically engineering eukaryotic cells and now considered as the major innovation in gene-related disorders. Nanotechnology has provided an alternate way to overcome the conventional problems where methods to deliver therapeutic agents have failed. The use of nanotechnology has the potential to safe-side the CRISPR/Cas9 components delivery by using customized polymeric nanoparticles for safety and efficacy. The pairing of two (CRISPR/Cas9 and nanotechnology) has the potential for opening new avenues in therapeutic use. In this review, we will discuss the most recent advances in developing nanoparticle-based CRISPR/Cas9 gene editing cargo delivery with a focus on several polymeric nanoparticles including fabrication proposals to combat microbial infections.
Collapse
Affiliation(s)
- Richa Verma
- Center for Nanobiotechnology Research, Department of Biological Sciences, Alabama State University, Montgomery, AL, 36104, USA
| | - Rajnish Sahu
- Center for Nanobiotechnology Research, Department of Biological Sciences, Alabama State University, Montgomery, AL, 36104, USA
| | - Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University, Jaipur, Rajasthan, 303002, India
| | - Timothy E Egbo
- Department of Biological Sciences, College of Science Technology Engineering and Mathematics, Alabama State University, Montgomery, AL, 36104, USA.
| |
Collapse
|
41
|
Di Pietro M, Filardo S, Romano S, Sessa R. Chlamydia trachomatis and Chlamydia pneumoniae Interaction with the Host: Latest Advances and Future Prospective. Microorganisms 2019; 7:microorganisms7050140. [PMID: 31100923 PMCID: PMC6560445 DOI: 10.3390/microorganisms7050140] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/07/2019] [Accepted: 05/14/2019] [Indexed: 12/27/2022] Open
Abstract
Research in Chlamydia trachomatis and Chlamydia pneumoniae has gained new traction due to recent advances in molecular biology, namely the widespread use of the metagenomic analysis and the development of a stable genomic transformation system, resulting in a better understanding of Chlamydia pathogenesis. C. trachomatis, the leading cause of bacterial sexually transmitted diseases, is responsible of cervicitis and urethritis, and C. pneumoniae, a widespread respiratory pathogen, has long been associated with several chronic inflammatory diseases with great impact on public health. The present review summarizes the current evidence regarding the complex interplay between C. trachomatis and host defense factors in the genital micro-environment as well as the key findings in chronic inflammatory diseases associated to C. pneumoniae.
Collapse
Affiliation(s)
- Marisa Di Pietro
- Department of Public Health and Infectious Diseases, Section of Microbiology, University of Rome "Sapienza", 00185 Rome, Italy.
| | - Simone Filardo
- Department of Public Health and Infectious Diseases, Section of Microbiology, University of Rome "Sapienza", 00185 Rome, Italy.
| | - Silvio Romano
- Cardiology, Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy.
| | - Rosa Sessa
- Department of Public Health and Infectious Diseases, Section of Microbiology, University of Rome "Sapienza", 00185 Rome, Italy.
| |
Collapse
|
42
|
Lv X, Li Z, Guan J, Zhang J, Xu B, He W, Lan Y, Zhao K, Lu H, Song D, Gao F. ATN-161 reduces virus proliferation in PHEV-infected mice by inhibiting the integrin α5β1-FAK signaling pathway. Vet Microbiol 2019; 233:147-153. [PMID: 31176401 DOI: 10.1016/j.vetmic.2019.04.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 04/24/2019] [Accepted: 04/24/2019] [Indexed: 02/08/2023]
Abstract
Porcine hemagglutinating encephalomyelitis virus (PHEV) is a typical neurotropic virus that can cause obvious nerve damage. Integrin α5β1 is a transmembrane macromolecular that closely related to neurological function. We recently demonstrated that integrin α5β1 plays a critical role in PHEV invasion in vitro. To determine the function and mechanism of integrin α5β1 in virus proliferation in vivo, we established a mouse model of PHEV infection. Integrin α5β1-FAK signaling pathway was activated in PHEV-infected mice by qPCR, Western blotting, and GST pull-down assays. Viral proliferation and integrin α5β1-FAK signaling pathway were significantly inhibited after intravenous injection of ATN-161, an integrin α5β1 inhibitor. Through a histological analysis, we found that ATN-161-treated mice only showed pathological changes in neuronal cytoplasmic swelling at 5 day post-infection. In summary, our results provide the first evidence that ATN-161 inhibits the proliferation of PHEV in mice and explores its underlying mechanisms of action.
Collapse
Affiliation(s)
- Xiaoling Lv
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zi Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jiyu Guan
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Jing Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Baofeng Xu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Wenqi He
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yungang Lan
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Kui Zhao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Huijun Lu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun 130062, China
| | - Deguang Song
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Feng Gao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|