1
|
Gousy-Leblanc M, Merkling T, Colston-Nepali L, Lachance Linklater E, Elliott KH, Friesen VL. Differences between mates at the TLR1Lb locus are associated with lower reproductive success in a long-lived seabird. Sci Rep 2024; 14:31608. [PMID: 39738121 DOI: 10.1038/s41598-024-77750-7] [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: 12/14/2023] [Accepted: 10/24/2024] [Indexed: 01/01/2025] Open
Abstract
Genetic diversity can influence fitness components such as survival and reproductive success. Yet the association between genetic diversity and fitness based on neutral loci is sometime very weak and inconsistent, with relationships varying among taxa due to confounding effects of population demography and life history. Fitness-diversity relationships are likely to be stronger and more consistent for genes known to influence phenotypic traits, such as immunity-related genes, and may also depend on the genetic differences between breeding partners. We recorded breeding success of individuals and breeding pairs over 20 years to evaluate the relationships between reproductive success and both neutral genetic variation (using 7,830 single nucleotide polymorphisms) and functional variation (four toll-like receptor [TLRs] loci) with reproductive success in thick-billed murres (Uria lomvia). Individual genetic diversity (both neutral and functional) was unrelated to reproductive success, but surprisingly, successful multi-year reproductive success decreased with the genetic difference between breeding partners at TLR1Lb. This result may be due to an advantage of specific alleles at TLR1Lb. This study is one of few addressing both individual genetic variation and genetic similarity between mates at both neutral and functional variation in a long-lived bird.
Collapse
Affiliation(s)
- Marianne Gousy-Leblanc
- Department of Natural Resource Sciences, McGill University, Ste-Anne-de-Bellevue, QC, Canada.
| | - Thomas Merkling
- Department of Natural Resource Sciences, McGill University, Ste-Anne-de-Bellevue, QC, Canada
| | | | | | - Kyle H Elliott
- Department of Natural Resource Sciences, McGill University, Ste-Anne-de-Bellevue, QC, Canada
| | - Vicki L Friesen
- Department of Biology, Queen's University, Kingston, ON, Canada
| |
Collapse
|
2
|
Broder KC, Matrosova VY, Tkavc R, Gaidamakova EK, Ho LTVT, Macintyre AN, Soc A, Diallo A, Darnell SC, Bash S, Daly MJ, Jerse AE, Liechti GW. Irradiated whole cell Chlamydia vaccine confers significant protection in a murine genital tract challenge model. NPJ Vaccines 2024; 9:207. [PMID: 39528548 PMCID: PMC11554809 DOI: 10.1038/s41541-024-00968-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 09/11/2024] [Indexed: 11/16/2024] Open
Abstract
Chlamydia trachomatis infections are the most common bacterial STIs globally and can lead to serious morbidity if untreated. Development of a killed, whole-cell vaccine has been stymied by coincident epitope destruction during inactivation. Here, we present a prototype Chlamydia vaccine composed of elementary bodies (EBs) from the related mouse pathogen, Chlamydia muridarum (Cm). EBs inactivated by gamma rays (Ir-Cm) in the presence of the antioxidant Mn2+-Decapeptide (DEHGTAVMLK) Phosphate (MDP) are protected from epitope damage but not DNA damage. Cm EBs gamma-inactivated with MDP retain their structure and provide significant protection in a murine genital tract infection model. Mice vaccinated with Ir-Cm (+MDP) exhibited elevated levels of Cm-specific IgG and IgA antibodies, reduced bacterial burdens, accelerated clearance, and distinctive cytokine responses compared to unvaccinated controls and animals vaccinated with EBs irradiated without MDP. Preserving EB epitopes with MDP during gamma inactivation offers the potential for a polyvalent, whole-cell vaccine against C. trachomatis.
Collapse
Affiliation(s)
- Kieran C Broder
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Department of Microbiology, Immunology, and Pathology, College of Veterinary Medicine and Biological Sciences, Colorado State University, Ft. Collins, CO, USA
| | - Vera Y Matrosova
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Rok Tkavc
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Elena K Gaidamakova
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Lam Thuy Vi Tran Ho
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | | | - Anthony Soc
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Aissata Diallo
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Stephen C Darnell
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Sarah Bash
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Michael J Daly
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - Ann E Jerse
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| | - George W Liechti
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences, Bethesda, MD, USA.
| |
Collapse
|
3
|
Russi RC, del Balzo D, Reidel IG, Alonso Bivou M, Flor N, Lujan A, Sanchez D, Damiani MT, Veaute C. Evaluation of three formulations based on Polymorphic membrane protein D in mice infected with Chlamydia trachomatis. Front Immunol 2023; 14:1267684. [PMID: 38045697 PMCID: PMC10690417 DOI: 10.3389/fimmu.2023.1267684] [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: 07/26/2023] [Accepted: 10/23/2023] [Indexed: 12/05/2023] Open
Abstract
The significant impact of Chlamydia trachomatis(Ct) infections worldwide highlights the need to develop a prophylactic vaccine that elicits effective immunity and protects the host from the immunopathological effects of Ct infection. The aim of this study was to evaluate a vaccine based on a fragment of the Polymorphic membrane protein D (FPmpD) of C. trachomatis as an immunogen using a heterologous DNA prime-protein boost strategy in female mice Three different formulations were evaluated as protein boost: free recombinant FPmpD (rFPmpD) or rFPmpD formulated with a liposomal adjuvant alternatively supplemented with CpG or a cationic gemini lipopeptide as immunostimulants. The three candidates induced an increase in the cervicovaginal and systemic titers of anti-rFPmpD antibodies in two strains of mice (BALB/c and C57BL/6), with no evidence of fertility alterations. The three formulations induced a rapid and robust humoral immune response upon the Ct challenge. However, the booster with free rFPmpD more efficiently reduced the shedding of infective Ct and prevented the development of immunopathology. The formulations containing adjuvant induced a strong inflammatory reaction in the uterine tissue. Hence, the prime-boost strategy with the adjuvant-free FPmpD vaccine formulation might constitute a promissory candidate to prevent C. trachomatis intravaginal infection.
Collapse
Affiliation(s)
- Romina Cecilia Russi
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
- Experimental Immunology Laboratory, School of Biochemistry and Biological Sciences, National University of Litoral, Ciudad Universitaria, Santa Fe, Argentina
| | - Diego del Balzo
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Ivana Gabriela Reidel
- Experimental Immunology Laboratory, School of Biochemistry and Biological Sciences, National University of Litoral, Ciudad Universitaria, Santa Fe, Argentina
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Mariano Alonso Bivou
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Noelia Flor
- Experimental Immunology Laboratory, School of Biochemistry and Biological Sciences, National University of Litoral, Ciudad Universitaria, Santa Fe, Argentina
| | - Agustín Lujan
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Diego Sanchez
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Teresa Damiani
- Laboratorio de Bioquímica e Inmunidad, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Instituto de Medicina y Biología Experimental de Cuyo, Consejo Nacional de Investigaciones Científicas y Técnicas (IMBECUCONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Carolina Veaute
- Experimental Immunology Laboratory, School of Biochemistry and Biological Sciences, National University of Litoral, Ciudad Universitaria, Santa Fe, Argentina
| |
Collapse
|
4
|
Turman BJ, Darville T, O'Connell CM. Plasmid-mediated virulence in Chlamydia. Front Cell Infect Microbiol 2023; 13:1251135. [PMID: 37662000 PMCID: PMC10469868 DOI: 10.3389/fcimb.2023.1251135] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Chlamydia trachomatis infection of ocular conjunctiva can lead to blindness, while infection of the female genital tract can lead to chronic pelvic pain, ectopic pregnancy, and/or infertility. Conjunctival and fallopian tube inflammation and the resulting disease sequelae are attributed to immune responses induced by chlamydial infection at these mucosal sites. The conserved chlamydial plasmid has been implicated in enhancing infection, via improved host cell entry and exit, and accelerating innate inflammatory responses that lead to tissue damage. The chlamydial plasmid encodes eight open reading frames, three of which have been associated with virulence: a secreted protein, Pgp3, and putative transcriptional regulators, Pgp4 and Pgp5. Although Pgp3 is an important plasmid-encoded virulence factor, recent studies suggest that chlamydial plasmid-mediated virulence extends beyond the expression of Pgp3. In this review, we discuss studies of genital, ocular, and gastrointestinal infection with C. trachomatis or C. muridarum that shed light on the role of the plasmid in disease development, and the potential for tissue and species-specific differences in plasmid-mediated pathogenesis. We also review evidence that plasmid-associated inflammation can be independent of bacterial burden. The functions of each of the plasmid-encoded proteins and potential molecular mechanisms for their role(s) in chlamydial virulence are discussed. Although the understanding of plasmid-associated virulence has expanded within the last decade, many questions related to how and to what extent the plasmid influences chlamydial infectivity and inflammation remain unknown, particularly with respect to human infections. Elucidating the answers to these questions could improve our understanding of how chlamydia augment infection and inflammation to cause disease.
Collapse
Affiliation(s)
- Breanna J. Turman
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Toni Darville
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
- Department of Pediatrics, University of North Carolina, Chapel Hill, NC, United States
| | | |
Collapse
|
5
|
Ait Djebbara S, Mcheik S, Percier P, Segueni N, Poncelet A, Truyens C. The macrophage infectivity potentiator of Trypanosoma cruzi induces innate IFN-γ and TNF-α production by human neonatal and adult blood cells through TLR2/1 and TLR4. Front Immunol 2023; 14:1180900. [PMID: 37304288 PMCID: PMC10250606 DOI: 10.3389/fimmu.2023.1180900] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 05/10/2023] [Indexed: 06/13/2023] Open
Abstract
We previously identified the recombinant (r) macrophage (M) infectivity (I) potentiator (P) of the protozoan parasite Trypanosoma cruzi (Tc) (rTcMIP) as an immuno-stimulatory protein that induces the release of IFN-γ, CCL2 and CCL3 by human cord blood cells. These cytokines and chemokines are important to direct a type 1 adaptive immune response. rTcMIP also increased the Ab response and favored the production of the Th1-related isotype IgG2a in mouse models of neonatal vaccination, indicating that rTcMIP could be used as a vaccine adjuvant to enhance T and B cell responses. In the present study, we used cord and adult blood cells, and isolated NK cells and human monocytes to investigate the pathways and to decipher the mechanism of action of the recombinant rTcMIP. We found that rTcMIP engaged TLR1/2 and TLR4 independently of CD14 and activated the MyD88, but not the TRIF, pathway to induce IFN-γ production by IL-15-primed NK cells, and TNF-α secretion by monocytes and myeloid dendritic cells. Our results also indicated that TNF-α boosted IFN-γ expression. Though cord blood cells displayed lower responses than adult cells, our results allow to consider rTcMIP as a potential pro-type 1 adjuvant that might be associated to vaccines administered in early life or later.
Collapse
Affiliation(s)
- Sarra Ait Djebbara
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Saria Mcheik
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Pauline Percier
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Service Immune Response, Sciensano, Brussels, Belgium
| | - Noria Segueni
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Antoine Poncelet
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Carine Truyens
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
6
|
Intracellular lifestyle of Chlamydia trachomatis and host-pathogen interactions. Nat Rev Microbiol 2023:10.1038/s41579-023-00860-y. [PMID: 36788308 DOI: 10.1038/s41579-023-00860-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2023] [Indexed: 02/16/2023]
Abstract
In recent years, substantial progress has been made in the understanding of the intracellular lifestyle of Chlamydia trachomatis and how the bacteria establish themselves in the human host. As an obligate intracellular pathogenic bacterium with a strongly reduced coding capacity, C. trachomatis depends on the provision of nutrients from the host cell. In this Review, we summarize the current understanding of how C. trachomatis establishes its intracellular replication niche, how its metabolism functions in the host cell, how it can defend itself against the cell autonomous and innate immune response and how it overcomes adverse situations through the transition to a persistent state. In particular, we focus on those processes for which a mechanistic understanding has been achieved.
Collapse
|
7
|
Chan YT, Cheok YY, Cheong HC, Tan GMY, Seow SR, Tang TF, Sulaiman S, Looi CY, Gupta R, Arulanandam B, Wong WF. Influx of podoplanin-expressing inflammatory macrophages into the genital tract following Chlamydia infection. Immunol Cell Biol 2023; 101:305-320. [PMID: 36658328 DOI: 10.1111/imcb.12621] [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: 06/01/2022] [Revised: 12/25/2022] [Accepted: 01/17/2023] [Indexed: 01/21/2023]
Abstract
Genital Chlamydia trachomatis infection remains a major health issue as it causes severe complications including pelvic inflammatory disease, ectopic pregnancy and infertility in females as a result of infection-associated chronic inflammation. Podoplanin, a transmembrane receptor, has been previously reported on inflammatory macrophages. Thus, strategies that specifically target podoplanin might be able to reduce local inflammation. This study investigated the expression level and function of podoplanin in a C. trachomatis infection model. C57BL/6 mice infected with the mouse pathogen Chlamydia muridarum were examined intermittently from days 1 to 60 using flow cytometry analysis. Percentages of conventional macrophages (CD11b+ CD11c- F4/80+ ) versus inflammatory macrophages (CD11b+ CD11c+ F4/80+ ), and the expression of podoplanin in these cells were investigated. Subsequently, a podoplanin-knockout RAW264.7 cell was used to evaluate the function of podoplanin in C. trachomatis infection. Our findings demonstrated an increased CD11b+ cell volume in the spleen at day 9 after the infection, with augmented podoplanin expression, especially among the inflammatory macrophages. A large number of podoplanin-expressing macrophages were detected in the genital tract of C. muridarum-infected mice. Furthermore, analysis of the C. trachomatis-infected patients demonstrated a higher percentage of podoplanin-expressing monocytes than that in the noninfected controls. Using an in vitro infection in a transwell migration assay, we identified that macrophages deficient in podoplanin displayed defective migratory function toward C. trachomatis-infected HeLa 229 cells. Lastly, using immunoprecipitation-mass spectrometry method, we identified two potential podoplanin interacting proteins, namely, Cofilin 1 and Talin 1 actin-binding proteins. The present study reports a role of podoplanin in directing macrophage migration to the chlamydial infection site. Our results suggest a potential for reducing inflammation in individuals with chronic chlamydial infections by targeting podoplanin.
Collapse
Affiliation(s)
- Yee Teng Chan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yi Ying Cheok
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Heng Choon Cheong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Grace Min Yi Tan
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Shi Rui Seow
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Ting Fang Tang
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Sofiah Sulaiman
- Department of Obstetrics and Gynecology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Chung Yeng Looi
- School of Bioscience, Faculty of Health & Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Rishein Gupta
- Center of Excellence in Infection Genomics, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, USA
| | - Bernard Arulanandam
- Center of Excellence in Infection Genomics, South Texas Center for Emerging Infectious Diseases, University of Texas at San Antonio, San Antonio, TX, USA.,Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | - Won Fen Wong
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
Interplay between C1-inhibitor and group IIA secreted phospholipase A 2 impairs their respective function. Immunol Res 2023; 71:70-82. [PMID: 36385678 PMCID: PMC9845149 DOI: 10.1007/s12026-022-09331-7] [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: 07/22/2022] [Accepted: 10/14/2022] [Indexed: 11/18/2022]
Abstract
High levels of human group IIA secreted phospholipase A2 (hGIIA) have been associated with various inflammatory disease conditions. We have recently shown that hGIIA activity and concentration are increased in the plasma of patients with hereditary angioedema due to C1-inhibitor deficiency (C1-INH-HAE) and negatively correlate with C1-INH plasma activity. In this study, we analyzed whether the presence of both hGIIA and C1-INH impairs their respective function on immune cells. hGIIA, but not recombinant and plasma-derived C1-INH, stimulates the production of IL-6, CXCL8, and TNF-α from peripheral blood mononuclear cells (PBMCs). PBMC activation mediated by hGIIA is blocked by RO032107A, a specific hGIIA inhibitor. Interestingly, C1-INH inhibits the hGIIA-induced production of IL-6, TNF-α, and CXCL8, while it does not affect hGIIA enzymatic activity. On the other hand, hGIIA reduces the capacity of C1-INH at inhibiting C1-esterase activity. Spectroscopic and molecular docking studies suggest a possible interaction between hGIIA and C1-INH but further experiments are needed to confirm this hypothesis. Together, these results provide evidence for a new interplay between hGIIA and C1-INH, which may be important in the pathophysiology of hereditary angioedema.
Collapse
|
9
|
Radwanska M, de Lemos Esteves F, Linsen L, Coltel N, Cencig S, Widart J, Massart AC, Colson S, Di Paolo A, Percier P, Ait Djebbara S, Guillonneau F, Flamand V, De Pauw E, Frère JM, Carlier Y, Truyens C. Macrophage-infectivity potentiator of Trypanosoma cruzi (TcMIP) is a new pro-type 1 immuno-stimulating protein for neonatal human cells and vaccines in mice. Front Immunol 2023; 14:1138526. [PMID: 37033946 PMCID: PMC10077492 DOI: 10.3389/fimmu.2023.1138526] [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: 01/05/2023] [Accepted: 02/27/2023] [Indexed: 04/11/2023] Open
Abstract
This work identifies the protein "macrophage infectivity potentiator" of Trypanosoma cruzi trypomastigotes, as supporting a new property, namely a pro-type 1 immunostimulatory activity on neonatal cells. In its recombinant form (rTcMIP), this protein triggers the secretion of the chemokines CCL2 and CCL3 by human umbilical cord blood cells from healthy newborns, after 24h in vitro culture. Further stimulation for 72h results in secretion of IFN-γ, provided cultures are supplemented with IL-2 and IL-18. rTcMIP activity is totally abolished by protease treatment and is not associated with its peptidyl-prolyl cis-trans isomerase enzymatic activity. The ability of rTcMIP to act as adjuvant was studied in vivo in neonatal mouse immunization models, using acellular diphtheria-tetanus-pertussis-vaccine (DTPa) or ovalbumin, and compared to the classical alum adjuvant. As compared to the latter, rTcMIP increases the IgG antibody response towards several antigens meanwhile skewing antibody production towards the Th-1 dependent IgG2a isotype. The amplitude of the rTcMIP adjuvant effect varied depending on the antigen and the co-presence of alum. rTcMIP did by contrast not increase the IgE response to OVA combined with alum. The discovery of the rTcMIP immunostimulatory effect on neonatal cells opens new possibilities for potential use as pro-type 1 adjuvant for neonatal vaccines. This, in turn, may facilitate the development of more efficient vaccines that can be given at birth, reducing infection associated morbidity and mortality which are the highest in the first weeks after birth.
Collapse
Affiliation(s)
- Magdalena Radwanska
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Loes Linsen
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Nicolas Coltel
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Sabrina Cencig
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Joelle Widart
- Laboratory of Mass Spectrometry (LSM), Department of Chemistry, Université de Liège, Liège, Belgium
| | - Anne-Cécile Massart
- Laboratory of Mass Spectrometry (LSM), Department of Chemistry, Université de Liège, Liège, Belgium
| | - Séverine Colson
- Center for Protein Engineering (CIP), Université de Liège (ULg), Liège, Belgium
| | - Alexandre Di Paolo
- Center for Protein Engineering (CIP), Université de Liège (ULg), Liège, Belgium
| | - Pauline Percier
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Sarra Ait Djebbara
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - François Guillonneau
- Laboratory of Mass Spectrometry (LSM), Department of Chemistry, Université de Liège, Liège, Belgium
| | - Véronique Flamand
- Institute for Medical Immunology (IMI), and ULB Center for Research in Immunology (U-CRI), Gosselies, Belgium
| | - Edwin De Pauw
- Laboratory of Mass Spectrometry (LSM), Department of Chemistry, Université de Liège, Liège, Belgium
| | - Jean-Marie Frère
- Center for Protein Engineering (CIP), Université de Liège (ULg), Liège, Belgium
| | - Yves Carlier
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- Department of Tropical Medicine, School of Public Health and Tropical Medicine, Tulane University, New Orleans, MA, United States
| | - Carine Truyens
- Laboratory of Parasitology, Faculty of Medicine, and ULB Center for Research in Immunology (UCRI), Université Libre de Bruxelles (ULB), Brussels, Belgium
- *Correspondence: Carine Truyens,
| |
Collapse
|
10
|
Pal S, Sheff S, Al-Kuhlani M, Ojcius DM, de la Maza LM. Role of TRAIL-R in Primary and Secondary Genital and Respiratory Chlamydia muridarum Infections in Mice. Microbiol Spectr 2022; 10:e0161722. [PMID: 35876584 PMCID: PMC9431660 DOI: 10.1128/spectrum.01617-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/01/2022] [Indexed: 11/20/2022] Open
Abstract
The tumor necrosis factor (TNF)-related apoptosis-inducing ligand receptor (TRAIL-R) suppresses inflammation and could therefore affect the course of Chlamydia infections and their long-term sequelae. Wild-type (WT) and TRAIL-R-/- C57BL/6 mice were inoculated vaginally with Chlamydia muridarum; the course of the infection was followed with vaginal cultures and the presence of hydrosalpinx determined. To evaluate the role of TRAIL-R following a secondary infection, the mice were vaginally reinfected. WT and TRAIL-R-/- male mice were also infected and reinfected in the respiratory tract, and the course of the diseases and the infections were followed. Following the primary and secondary vaginal infection, no significant differences in vaginal shedding or hydrosalpinx formation were observed between the WT and TRAIL-R-/- mice. The WT and TRAIL-R-/- mice mounted antibody responses in serum and vaginal washes that were not significantly different. After the primary and secondary intranasal infections of the male mice, changes in body weight were determined, and no significant differences were observed between the WT and TRAIL-R-/- mice. Ten days after the primary and the secondary infections, the weight of the lungs and number of C. muridarum inclusion forming units (IFU) were determined. The lungs of the WT mice weighed less compared with the TRAIL-R-/- mice following a primary infection but not after a secondary infection. No differences in the number of C. muridarum IFU in the lungs were observed between the two groups of mice. In conclusion, despite playing a role in inflammation cell-signaling pathways in vitro, TRAIL-R does not appear to play a major role in the susceptibility, clinical outcomes, or long-term sequelae of C. muridarum infections in vivo. IMPORTANCE TNF-related apoptosis-inducing ligand receptor (TRAIL-R) is involved in suppressing inflammatory responses. Bacterial pathogens such as Chlamydia spp. elicit inflammatory responses in humans following genital, ocular, and respiratory infections. The inflammatory responses are important to control the spread of Chlamydia. However, in certain instances, these inflammatory responses can produce long-term sequelae, including fibrosis. Fibrosis, or scarring, in the genital tract, eye, and respiratory system results in functional deficiencies, including infertility, blindness, and chronic obstructive lung disease, respectively. The goal of this study was to determine if mice deficient in TRAIL-R infected in the genital and respiratory tracts with Chlamydia spp. suffer more or less severe infections, infertility, or lung diseases than wild-type mice. Our results show no differences between the immune responses, infection severity, and long-term sequelae between TRAIL-R knockout and wild-type animals following a genital or a respiratory infection with Chlamydia.
Collapse
Affiliation(s)
- Sukumar Pal
- Department of Pathology and Laboratory Medicine, Medical Sciences I, University of California, Irvine, Irvine, California, USA
| | - Sydni Sheff
- Department of Pathology and Laboratory Medicine, Medical Sciences I, University of California, Irvine, Irvine, California, USA
| | - Mufadhal Al-Kuhlani
- Department of Biomedical Sciences, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, California, USA
- Life Science Department, Fresno City College, Fresno, California, USA
| | - David M. Ojcius
- Department of Biomedical Sciences, Arthur Dugoni School of Dentistry, University of the Pacific, San Francisco, California, USA
| | - Luis M. de la Maza
- Department of Pathology and Laboratory Medicine, Medical Sciences I, University of California, Irvine, Irvine, California, USA
| |
Collapse
|
11
|
Marcella S, Apicella B, Secondo A, Palestra F, Opromolla G, Ciardi R, Tedeschi V, Ferrara AL, Russo C, Rosaria Galdiero M, Cristinziano L, Modestino L, Spadaro G, Fiorelli A, Loffredo S. Size-based effects of anthropogenic ultrafine particles on activation of human lung macrophages. ENVIRONMENT INTERNATIONAL 2022; 166:107395. [PMID: 35839670 DOI: 10.1016/j.envint.2022.107395] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/31/2022] [Accepted: 07/04/2022] [Indexed: 06/15/2023]
Abstract
The anthropogenic particulate matter (PM), suspended air dust that can be inhaled by humans and deposited in the lungs, is one of the main pollutants in the industrialized cities atmosphere. Recent studies have shown that PM has adverse effects on respiratory diseases. These effects are mainly due to the ultrafine particles (PM0.1, PM < 100 nm), which, thanks to their PM size, are efficiently deposited in nasal, tracheobronchial, and alveolar regions. Pulmonary macrophages are a heterogeneous cell population distributed in different lung compartments, whose role in inflammatory response to injury is of particular relevance. In this study, we investigated the effect of PM0.1 on Human Lung Macrophages (HLMs) activation evaluated as proinflammatory cytokines and chemokine release, Reactive Oxygen Species (ROS) production and intracellular Ca2+concentration ([Ca2+]i). Furthermore, PM0.1, after removal of organic fraction, was fractionated in nanoparticles both smaller (NP20) and bigger (NP100) than 20 nm by a properlydeveloped analytical protocol, allowed isolating their individual contribution. Interestingly, while PM0.1 and NP20 induced stimulatory effects on HLM cytokines release, NP100 had not effect. In particular, PM0.1 induced IL-6, IL-1β, TNF-α, but not CXCL8, release from HLMs. Moreover, PM0.1, NP20 and NP100 did not induce β-glucuronidase release, a preformed mediator contained in HLMs. The long time necessary for cytokines release (18 h) suggested that PM0.1 and NP20 could induce ex-novo production of the tested mediators. Accordingly, after 6 h of incubation, PM0.1 and NP20 induced mRNA expression of IL-6, TNF-α and IL-1β. Moreover, NP20 induced ROS production and [Ca2+]i increase in a time-dependent manner, without producing cytotoxicity. Collectively, the present data highlight the main proinflammatory role of NP20 among PM fractions. This is particularly of concern because this fraction is not currently covered by legal limits as it is not easily measured at the exhausts by the available technical methodologies, suggesting that it is mandatory to search for new monitoring techniques and strategies for limiting NP20 formation.
Collapse
Affiliation(s)
- Simone Marcella
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, 80131 Naples, Italy.
| | - Barbara Apicella
- Istituto di Scienze e Tecnologie per l'Energia e la Mobilità Sostenibili (STEMS)-CNR, 80125 Naples, Italy.
| | - Agnese Secondo
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy.
| | - Francesco Palestra
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, 80131 Naples, Italy.
| | - Giorgia Opromolla
- Translational Medical and Surgical Science, University of Campania Luigi Vanvitelli, 80131 Naples, Italy.
| | - Renato Ciardi
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, 80131 Naples, Italy.
| | - Valentina Tedeschi
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, University of Naples Federico II, 80131 Naples, Italy.
| | - Anne Lise Ferrara
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy.
| | - Carmela Russo
- Istituto di Scienze e Tecnologie per l'Energia e la Mobilità Sostenibili (STEMS)-CNR, 80125 Naples, Italy.
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy.
| | - Leonardo Cristinziano
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, 80131 Naples, Italy.
| | - Luca Modestino
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, 80131 Naples, Italy.
| | - Giuseppe Spadaro
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, 80131 Naples, Italy.
| | - Alfonso Fiorelli
- Translational Medical and Surgical Science, University of Campania Luigi Vanvitelli, 80131 Naples, Italy.
| | - Stefania Loffredo
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, WAO Center of Excellence, 80131 Naples, Italy; Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council, 80131 Naples, Italy.
| |
Collapse
|
12
|
Tsujikawa R, Thapa J, Okubo T, Nakamura S, Zhang S, Furuta Y, Higashi H, Yamaguchi H. Chlamydia trachomatis L2/434/Bu Favors Hypoxia for its Growth in Human Lymphoid Jurkat Cells While Maintaining Production of Proinflammatory Cytokines. Curr Microbiol 2022; 79:265. [PMID: 35859064 DOI: 10.1007/s00284-022-02961-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 06/27/2022] [Indexed: 11/03/2022]
Abstract
The role of lymphocytes as a cornerstone of the inflammatory response in the invasive pathogenesis of Chlamydia trachomatis (Ct) LGV (L1-3) infection is unclear. Therefore, we assessed whether the adaptation of CtL2 to immortal lymphoid Jurkat cells under hypoxic conditions occurred through proinflammatory cytokine profile modification. The quantities of inclusion-forming units with chlamydial 16S rDNA confirmed that CtL2 grew well under hypoxic rather than normoxic conditions in the cells. Confocal microscopic imaging and transmission electron microscopy revealed the presence of bacterial progeny in the inclusions and showed that the inclusions were larger under hypoxic rather than normoxic conditions; this was supported by the results of 3D image construction. Furthermore, PCR-based analysis of proinflammatory cytokines revealed that the gene expression levels under hypoxic conditions were significantly higher than those under normoxic conditions. In particular, the expression of two genes (CXCL8 and CXCR3) was significantly diminished under normoxic conditions. Taken together, the results indicated that hypoxia promoted CtL2 growth in Jurkat cells while maintaining the levels of proinflammatory cytokines. Thus, Ct LGV infection in lymphocytes under hypoxic conditions might be crucial to a complete understanding of the invasive pathogenesis.
Collapse
Affiliation(s)
- Ryoya Tsujikawa
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kitaku, Sapporo, Hokkaido, 060-0812, Japan
| | - Jeewan Thapa
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, North-20, West-10, Kita-ku, Sapporo, 001-0020, Japan
| | - Torahiko Okubo
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kitaku, Sapporo, Hokkaido, 060-0812, Japan
| | - Shinji Nakamura
- Division of Biomedical Imaging Research, and Division of Ultrastructural Research, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Saicheng Zhang
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kitaku, Sapporo, Hokkaido, 060-0812, Japan
| | - Yoshikazu Furuta
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, North-20, West-10, Kita-ku, Sapporo, 001-0020, Japan
| | - Hideaki Higashi
- Division of Infection and Immunity, International Institute for Zoonosis Control, Hokkaido University, North-20, West-10, Kita-ku, Sapporo, 001-0020, Japan
| | - Hiroyuki Yamaguchi
- Department of Medical Laboratory Science, Faculty of Health Sciences, Hokkaido University, Kita 12, Nishi 5, Kitaku, Sapporo, Hokkaido, 060-0812, Japan.
| |
Collapse
|
13
|
Barnes AB, Keener RM, Schott BH, Wang L, Valdivia RH, Ko DC. Human genetic diversity regulating the TLR10/TLR1/TLR6 locus confers increased cytokines in response to Chlamydia trachomatis. HGG ADVANCES 2022; 3:100071. [PMID: 35047856 PMCID: PMC8756536 DOI: 10.1016/j.xhgg.2021.100071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 11/15/2021] [Indexed: 12/25/2022] Open
Abstract
Human genetic diversity can have profound effects on health outcomes upon exposure to infectious agents. For infections with Chlamydia trachomatis (C. trachomatis), the wide range of genital and ocular disease manifestations are likely influenced by human genetic differences that regulate interactions between C. trachomatis and host cells. We leveraged this diversity in cellular responses to demonstrate the importance of variation at the Toll-like receptor 1 (TLR1), TLR6, and TLR10 locus to cytokine production in response to C. trachomatis. We determined that a single-nucleotide polymorphism (SNP) (rs1057807), located in a region that forms a loop with the TLR6 promoter, is associated with increased expression of TLR1, TLR6, and TLR10 and secreted levels of ten C. trachomatis-induced cytokines. Production of these C. trachomatis-induced cytokines is primarily dependent on MyD88 and TLR6 based on experiments using inhibitors, blocking antibodies, RNAi, and protein overexpression. Population genetic analyses further demonstrated that the mean IL-6 response of cells from two European populations were higher than the mean response of cells from three African populations and that this difference was partially attributable to variation in rs1057807 allele frequency. In contrast, a SNP associated with a different pro-inflammatory cytokine (rs2869462 associated with the chemokine CXCL10) exhibited an opposite response, underscoring the complexity of how different genetic variants contribute to an individual's immune response. This multidisciplinary study has identified a long-range chromatin interaction and genetic variation that regulates TLR6 to broaden our understanding of how human genetic variation affects the C. trachomatis-induced immune response.
Collapse
Affiliation(s)
- Alyson B. Barnes
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Rachel M. Keener
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| | - Benjamin H. Schott
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
| | - Liuyang Wang
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Raphael H. Valdivia
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
| | - Dennis C. Ko
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC 27710, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC 27710, USA
- Division of Infectious Diseases, Department of Medicine, School of Medicine, Duke University, Durham, NC 27710, USA
| |
Collapse
|
14
|
Ling H, Luo L, Dai X, Chen H. Fallopian tubal infertility: the result of Chlamydia trachomatis-induced fallopian tubal fibrosis. Mol Cell Biochem 2021; 477:205-212. [PMID: 34652537 DOI: 10.1007/s11010-021-04270-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/29/2021] [Indexed: 11/26/2022]
Abstract
Chlamydia trachomatis is one of the most common pathogens of sexually transmitted diseases, and its incidence in genital tract infections is now 4.7% in south China. Infertility is the end result of C. trachomatis-induced fallopian tubal fibrosis and is receiving intense attention from scientists worldwide. To reduce the incidence of infertility, it is important to understand the pathology-related changes of the genital tract where C. trachomatis infection is significant, especially the mechanism of fibrosis formation. During fibrosis development, the fallopian tube becomes sticky and occluded, which will eventually lead to tubal infertility. At present, the mechanism of fallopian tubal fibrosis induced by C. trachomatis infection is unclear. Our study attempted to summarize the possible mechanisms of fibrosis caused by C. trachomatis infection in the fallopian tube by reviewing published studies and further providing potential therapeutic targets to reduce the occurrence of infertility. This study also provides ideas for future research. Factors leading to fallopian tube fibrosis include inflammatory factors, miRNA, ECT, cHSP, and host factors. We hypothesized that C. trachomatis mediates the transcription and translation of EMT and ECM via upregulating TGF signaling pathway, which leads to the formation of fallopian tube fibrosis and ultimately to tubal infertility.
Collapse
Affiliation(s)
- Hua Ling
- The First People's Hospital of Chenzhou, The First School of Clinical Medicine, Southern Medical University, Chenzhou, 423000, People's Republic of China
| | - Lipei Luo
- The First People's Hospital of Chenzhou, The First School of Clinical Medicine, Southern Medical University, Chenzhou, 423000, People's Republic of China
| | - Xingui Dai
- The First People's Hospital of Chenzhou, The First School of Clinical Medicine, Southern Medical University, Chenzhou, 423000, People's Republic of China.
- The First People's Hospital of Chenzhou, Chenzhou, 423000, People's Republic of China.
- The First Affiliated Hospital of Xiangnan University, Chenzhou, 423000, People's Republic of China.
| | - Hongliang Chen
- The First People's Hospital of Chenzhou, The First School of Clinical Medicine, Southern Medical University, Chenzhou, 423000, People's Republic of China.
- The First People's Hospital of Chenzhou, Chenzhou, 423000, People's Republic of China.
- The First Affiliated Hospital of Xiangnan University, Chenzhou, 423000, People's Republic of China.
| |
Collapse
|
15
|
Li DK, Feng HH, Mu YT, Yu JQ, Yang F. Extraction and bioinformatics analysis of Chlamydia trachomatis LpxA. Int Ophthalmol 2020; 41:667-673. [PMID: 33078228 DOI: 10.1007/s10792-020-01623-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: 04/06/2020] [Accepted: 10/07/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE The aim of this study is to clone the LpxA gene of Chlamydia trachomatis and analyze its biological characteristics. METHODS Specific primers were designed according to the sequence of Ct LpxA gene. LpxA gene was amplified by PCR and connected to pMD18-T vectors. Positive clones were selected for PCR and DNA sequencing. Finally, bioinformatics software was used to analyze the biological properties of LpxA protein. RESULTS The total length of LpxA gene was 840 bp, encoding 280 amino acids. LpxA protein has no signal peptide and was located in bacterial cytoplasm. The prediction of secondary structure showed that the α-helix, extended strand, β-turn and random coil accounted for 19.6%, 32.8%, 11.4% and 36%, respectively. According to the prediction of tertiary structure, three identical LpxA molecules constituted homologous trimers. It was predicted that there were 11 B cell epitopes in LpxA. CONCLUSION Ct Lpxa gene was cloned, and LpxA protein structure and function were predicted.
Collapse
Affiliation(s)
- De-Kun Li
- Department of Ophthalmology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Huan-Huan Feng
- Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, Hubei, China
| | - Ying-Tao Mu
- Department of TCM, Renmin Hospital, Hubei University of Medicine, No. 39 Middle Chaoyang Road, Shiyan, 442000, Hubei, China.
| | - Jin-Qiang Yu
- Department of Ophthalmology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Fang Yang
- Department of Ophthalmology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
16
|
Duncan SA, Sahu R, Dixit S, Singh SR, Dennis VA. Suppressors of Cytokine Signaling (SOCS)1 and SOCS3 Proteins Are Mediators of Interleukin-10 Modulation of Inflammatory Responses Induced by Chlamydia muridarum and Its Major Outer Membrane Protein (MOMP) in Mouse J774 Macrophages. Mediators Inflamm 2020; 2020:7461742. [PMID: 32684836 PMCID: PMC7333066 DOI: 10.1155/2020/7461742] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/10/2020] [Indexed: 12/26/2022] Open
Abstract
The immunopathology of chlamydial diseases is exacerbated by a broad-spectrum of inflammatory mediators, which we reported are inhibited by IL-10 in macrophages. However, the chlamydial protein moiety that induces the inflammatory mediators and the mechanisms by which IL-10 inhibits them are unknown. We hypothesized that Chlamydia major outer membrane protein (MOMP) mediates its disease pathogenesis, and the suppressor of cytokine signaling (SOCS)1 and SOCS3 proteins are mediators of the IL-10 inhibitory actions. Our hypothesis was tested by exposing mouse J774 macrophages to chlamydial stimulants (live Chlamydia muridarum and MOMP) with and without IL-10. MOMP significantly induced several inflammatory mediators (IL-6, IL-12p40, CCL5, CXCL10), which were dose-dependently inhibited by IL-10. Chlamydial stimulants induced the mRNA gene transcripts and protein expression of SOCS1 and SOCS3, with more SOCS3 expression. Notably, IL-10 reciprocally regulated their expression by reducing SOCS1 and increasing SOCS3. Specific inhibitions of MAPK pathways revealed that p38, JNK, and MEK1/2 are required for inducing inflammatory mediators as well as SOCS1 and SOCS3. Chlamydial stimulants triggered an M1 pro-inflammatory phenotype evidently by an enhanced nos2 (M1 marker) expression, which was skewed by IL-10 towards a more M2 anti-inflammatory phenotype by the increased expression of mrc1 and arg1 (M2 markers) and the reduced SOCS1/SOCS3 ratios. Neutralization of endogenously produced IL-10 augmented the secretion of inflammatory mediators, reduced SOCS3 expression, and skewed the chlamydial M1 to an M2 phenotype. Inhibition of proteasome degradation increased TNF but decreased IL-10, CCL5, and CXCL10 secretion by suppressing SOCS1 and SOCS3 expressions and dysregulating their STAT1 and STAT3 transcription factors. Our data show that SOCS1 and SOCS3 are regulators of IL-10 inhibitory actions, and underscore SOCS proteins as therapeutic targets for IL-10 control of inflammation for Chlamydia and other bacterial inflammatory diseases.
Collapse
Affiliation(s)
- Skyla A. Duncan
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| | - Rajnish Sahu
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| | - Saurabh Dixit
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| | - Shree R. Singh
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| | - Vida A. Dennis
- Center for NanoBiotechnology Research (CNBR), Department of Biological Sciences, Alabama State University, 1627 Harris Way, Montgomery, AL 36104, USA
| |
Collapse
|
17
|
Chlamydia psittaci PmpD-N Exacerbated Chicken Macrophage Function by Triggering Th2 Polarization and the TLR2/MyD88/NF-κB Signaling Pathway. Int J Mol Sci 2020; 21:ijms21062003. [PMID: 32183481 PMCID: PMC7139469 DOI: 10.3390/ijms21062003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 02/29/2020] [Accepted: 03/12/2020] [Indexed: 12/25/2022] Open
Abstract
The polymorphic membrane protein D (PmpD) is a highly conserved outer membrane protein which plays an important role in pathogenesis during Chlamydia psittaci infection. In this study, we evaluated the ability of the N-terminus of PmpD (PmpD-N) to modulate the functions of chicken macrophages and the signaling pathway(s) involved in PmpD-N-induced Toll-like receptors (TLRs), as well as interleukin (IL)-6 and IL-10 cytokine secretions. Thus, HD11 macrophages were treated with exogenous and intracellular PmpD-N of C. psittaci. The chlamydial growth was evaluated by enumeration of chlamydial loads in the infected macrophages. The phagocytic function of macrophages following PmpD-N treatment was detected by fluorescein-labeled Escherichia coli (E. coli). The concentration of nitric oxide (NO) secreted by HD11 macrophages was measured by the amount of NO2- in the culture supernatant using the Griess method. The cytokine secretions were assessed using multiplex cytokine ELISA kits. Expression levels of TLRs, myeloid differentiation factor 88 (MyD88), and nuclear factor kappa B (NF-κB) were analyzed by a Western blotting assay, as well as a luciferase assay, while NF-κB p65 nuclear translocation was assessed by confocal microscopy. The nuclear translocation of the transcription factor NF-κB was confirmed by evaluating its ability to combine with the corresponding promoter using the electrophoretic mobility shift assay (EMSA). After treatment with exogenous or endogenous PmpD-N, chlamydial loads and phagocytic functions were reduced significantly compared with those of the plasmid vector group, while NO secretions were reduced significantly compared with those of the lipopolysaccharide (LPS) treatment. Stimulation of HD11 cells with PmpD-N provoked the secretion of the Th2 cytokines, IL-6, and IL-10 and upregulated the expression of TLR2, TLR4, MyD88, and NF-κB. Furthermore, inhibition of TLR2, MyD88, and NF-κB in HD11 cells significantly decreased IL-6 and IL-10 cytokine levels, while NO production and phagocytosis increased significantly, strongly suggesting their involvement in PmpD-N-induced Th2 cytokine secretion and macrophage dysfunction. Our data indicate that C. psittaci PmpD-N inhibited macrophage functions by activating the Th2 immune response and the TLR2/MyD88/NF-κB signaling pathway.
Collapse
|
18
|
Chen H, Wen Y, Li Z. Clear Victory for Chlamydia: The Subversion of Host Innate Immunity. Front Microbiol 2019; 10:1412. [PMID: 31333596 PMCID: PMC6619438 DOI: 10.3389/fmicb.2019.01412] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 06/05/2019] [Indexed: 12/14/2022] Open
Abstract
As obligate intracellular bacterial pathogens, members of the Chlamydia genera are the pivotal triggers for a wide range of infections, which can lead to blinding trachoma, pelvic inflammation, and respiratory diseases. Because of their restricted parasitism inside eukaryotic cells, the pathogens have to develop multiple strategies for adaptation with the hostile intracellular environment—intrinsically present in all host cells—to survive. The strategies that are brought into play at different stages of chlamydial development mainly involve interfering with diverse innate immune responses, such as innate immune recognition, inflammation, apoptosis, autophagy, as well as the manipulation of innate immune cells to serve as potential niches for chlamydial replication. This review will focus on the innate immune responses against chlamydial infection, highlighting the underlying molecular mechanisms used by the Chlamydia spp. to counteract host innate immune defenses. Insights into these subtle pathogenic mechanisms not only provide a rationale for the augmentation of immune responses against chlamydial infection but also open avenues for further investigation of the molecular mechanisms driving the survival of these clinically important pathogens in host innate immunity.
Collapse
Affiliation(s)
- Hongliang Chen
- 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.,Department of Clinical Microbiology Laboratory, Chenzhou No. 1 People's Hospital, Chenzhou, 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
| | - 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
|
19
|
Lausen M, Christiansen G, Bouet Guldbæk Poulsen T, Birkelund S. Immunobiology of monocytes and macrophages during Chlamydia trachomatis infection. Microbes Infect 2018; 21:73-84. [PMID: 30528899 DOI: 10.1016/j.micinf.2018.10.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 10/11/2018] [Accepted: 10/11/2018] [Indexed: 02/03/2023]
Abstract
Infections caused by the intracellular bacterium Chlamydia trachomatis are a global health burden affecting more than 100 million people annually causing damaging long-lasting infections. In this review, we will present and discuss important aspects of the interaction between C. trachomatis and monocytes/macrophages.
Collapse
Affiliation(s)
- Mads Lausen
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220 Aalborg Ø, Denmark.
| | - Gunna Christiansen
- Department of Biomedicine, Aarhus University, Wilhelms Meyers Allé 4, 8000 Aarhus, Denmark
| | | | - Svend Birkelund
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220 Aalborg Ø, Denmark
| |
Collapse
|
20
|
Sun HS, Liu DX, Bai YY, Hu NW. Disease-association of different killer cell immunoglobulin-like receptors (KIR) and HLA-C gene combinations in reactive arthritis. Mod Rheumatol 2018; 29:531-537. [PMID: 29848119 DOI: 10.1080/14397595.2018.1483292] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Hong Sheng Sun
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Dong Xia Liu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Yan Yan Bai
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Nai Wen Hu
- Department of Rheumatology and Immunology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| |
Collapse
|
21
|
Mackern-Oberti JP, Motrich RD, Damiani MT, Saka HA, Quintero CA, Sánchez LR, Moreno-Sosa T, Olivera C, Cuffini C, Rivero VE. Male genital tract immune response against Chlamydia trachomatis infection. Reproduction 2018; 154:R99-R110. [PMID: 28878094 DOI: 10.1530/rep-16-0561] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 07/01/2017] [Accepted: 07/11/2017] [Indexed: 01/01/2023]
Abstract
Chlamydia trachomatis is the most commonly reported agent of sexually transmitted bacterial infections worldwide. This pathogen frequently leads to persistent, long-term, subclinical infections, which in turn may cause severe pathology in susceptible hosts. This is in part due to the strategies that Chlamydia trachomatis uses to survive within epithelial cells and to evade the host immune response, such as subverting intracellular trafficking, interfering signaling pathways and preventing apoptosis. Innate immune receptors such as toll-like receptors expressed on epithelial and immune cells in the genital tract mediate the recognition of chlamydial molecular patterns. After bacterial recognition, a subset of pro-inflammatory cytokines and chemokines are continuously released by epithelial cells. The innate immune response is followed by the initiation of the adaptive response against Chlamydia trachomatis, which in turn may result in T helper 1-mediated protection or in T helper 2-mediated immunopathology. Understanding the molecular mechanisms developed by Chlamydia trachomatis to avoid killing and host immune response would be crucial for designing new therapeutic approaches and developing protective vaccines. In this review, we focus on chlamydial survival strategies and the elicited immune responses in male genital tract infections.
Collapse
Affiliation(s)
- Juan Pablo Mackern-Oberti
- Instituto de Medicina y Biología Experimental de Cuyo. IMBECU-CONICETMendoza, Argentina .,Instituto de Fisiología. Facultad de Ciencias MédicasUniversidad Nacional de Cuyo, Mendoza, Argentina
| | - Rubén Darío Motrich
- Centro de Investigaciones en Bioquímica Clínica e Inmunología CIBICI-CONICETDepartamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Maria Teresa Damiani
- Instituto de Histología y Embriología de Mendoza. IHEM-CONICETFacultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Héctor Alex Saka
- Centro de Investigaciones en Bioquímica Clínica e Inmunología CIBICI-CONICETDepartamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | - Leonardo Rodolfo Sánchez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología CIBICI-CONICETDepartamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Tamara Moreno-Sosa
- Instituto de Medicina y Biología Experimental de Cuyo. IMBECU-CONICETMendoza, Argentina
| | - Carolina Olivera
- Centro de Investigaciones en Bioquímica Clínica e Inmunología CIBICI-CONICETDepartamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Cecilia Cuffini
- Instituto de Virología Dr. J. M. VanellaFacultad de Ciencias Médicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Virginia Elena Rivero
- Centro de Investigaciones en Bioquímica Clínica e Inmunología CIBICI-CONICETDepartamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
22
|
Lausen M, Christiansen G, Karred N, Winther R, Poulsen TBG, Palarasah Y, Birkelund S. Complement C3 opsonization of Chlamydia trachomatis facilitates uptake in human monocytes. Microbes Infect 2018; 20:328-336. [PMID: 29729435 DOI: 10.1016/j.micinf.2018.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 03/13/2018] [Accepted: 04/23/2018] [Indexed: 01/08/2023]
Abstract
Chlamydia trachomatis is an obligate intracellular bacterium that causes severe infections, which can lead to infertility and ectopic pregnancy. Although both innate and adaptive immune responses are elicited during chlamydial infection the bacterium succeeds to evade host defense mechanisms establishing chronic infections. Thus, studying the host-pathogen interaction during chlamydial infection is of importance to understand how C. trachomatis can cause chronic infections. Both the complement system and monocytes play essential roles in anti-bacterial defense, and, therefore, we investigated the interaction between the complement system and the human pathogens C. trachomatis D and L2. Complement competent serum facilitated rapid uptake of both chlamydial serovars into monocytes. Using immunoelectron microscopy, we showed that products of complement C3 were loosely deposited on the bacterial surface in complement competent serum and further characterization demonstrated that the deposited C3 product was the opsonin iC3b. Using C3-depleted serum we confirmed that complement C3 facilitates rapid uptake of chlamydiae into monocytes in complement competent serum. Complement facilitated uptake did not influence intracellular survival of C. trachomatis or C. trachomatis-induced cytokine secretion. Hence, C. trachomatis D and L2 activate the complement system leading to chlamydial opsonization by iC3b and subsequent phagocytosis, activation and bacterial elimination by human monocytes.
Collapse
Affiliation(s)
- Mads Lausen
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark
| | - Gunna Christiansen
- Department of Biomedicine, Aarhus University, Wilhelms Meyers Allé 4, 8000, Aarhus, Denmark
| | - Nichlas Karred
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark
| | - Robert Winther
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark
| | - Thomas Bouet Guldbæk Poulsen
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark
| | - Yaseelan Palarasah
- Unit for Thrombosis Research, Institute of Public Health, University of Southern Denmark, Esbjerg, Denmark
| | - Svend Birkelund
- Department of Health Science and Technology, Aalborg University, Fredrik Bajers Vej 3b, 9220, Aalborg Ø, Denmark.
| |
Collapse
|
23
|
Update on Chlamydia trachomatis Vaccinology. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00543-16. [PMID: 28228394 DOI: 10.1128/cvi.00543-16] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Attempts to produce a vaccine to protect against Chlamydia trachomatis-induced trachoma were initiated more than 100 years ago and continued for several decades. Using whole organisms, protective responses were obtained. However, upon exposure to C. trachomatis, disease exacerbation developed in some immunized individuals, precluding the implementation of the vaccine. Evidence of the role of C. trachomatis as a sexually transmitted pathogen started to emerge in the 1960s, and it soon became evident that it can cause acute infections and long-term sequelae in women, men, and newborns. The main focus of this minireview is to summarize recent findings and discuss formulations, including antigens, adjuvants, routes, and delivery systems for immunization, primarily explored in the female mouse model, with the goal of implementing a vaccine against C. trachomatis genital infections.
Collapse
|
24
|
Wang Y, Liu Q, Chen D, Guan J, Ma L, Zhong G, Shu H, Wu X. Chlamydial Lipoproteins Stimulate Toll-Like Receptors 1/2 Mediated Inflammatory Responses through MyD88-Dependent Pathway. Front Microbiol 2017; 8:78. [PMID: 28184217 PMCID: PMC5266682 DOI: 10.3389/fmicb.2017.00078] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/11/2017] [Indexed: 01/12/2023] Open
Abstract
Chlamydiae are very important pathogens which could cause several types of diseases in human, but little is known about its pathogenic mechanism. In order to elucidate host inflammatory response and the signal pathway induced by Chlamydial lipoproteins, the predicted lipoproteins of Chlamydia trachomatis were tested for their ability to induce the release of proinflammatory cytokines by mouse macrophages or human TLR (Toll-Like Receptor) expressing cell lines. The results showed that recombinant proteins of C. trachomatis D381, D541, D067, and D775 displayed a strong ability to induce the release of IL-8 in TLR expressing cell line. The signal pathways involved TLR1/2 and TLR2/CD14 but not TLR4. Moreover, except D067, the proinflammatory cytokine induction by D381, D541, and D775 required the thioacylation site (cysteine) for lipid modification and the induction was through MyD88-mediated pathway. Our data supported that lipoproteins played a vital role in pathogenesis of C. trachomatis-induced inflammatory responses via TLR pathway. It was the first study to characterize other chlamydial lipoproteins after identifying the role of MIP (D541) on pathogenesis of Chlamydial diseases.
Collapse
Affiliation(s)
- Yong Wang
- School of Basic Medical Sciences, Xiangya School of Medicine, Central South University Changsha, China
| | - Qiong Liu
- Department of Cardiovascular Diseases, Xiangya Hospital, Central South University Changsha, China
| | - Ding Chen
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | - Jie Guan
- School of Basic Medical Sciences, Xiangya School of Medicine, Central South University Changsha, China
| | - Linghui Ma
- School of Basic Medical Sciences, Xiangya School of Medicine, Central South University Changsha, China
| | - Guangming Zhong
- Department of Microbiology and Immunology, University of Texas Health Science Center at San Antonio San Antonio, TX, USA
| | - Hengping Shu
- School of Basic Medical Sciences, Xiangya School of Medicine, Central South University Changsha, China
| | - Xiang Wu
- School of Basic Medical Sciences, Xiangya School of Medicine, Central South University Changsha, China
| |
Collapse
|
25
|
Finethy R, Coers J. Sensing the enemy, containing the threat: cell-autonomous immunity to Chlamydia trachomatis. FEMS Microbiol Rev 2016; 40:875-893. [PMID: 28201690 PMCID: PMC5975928 DOI: 10.1093/femsre/fuw027] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/31/2016] [Accepted: 07/01/2016] [Indexed: 01/01/2023] Open
Abstract
The bacterium Chlamydia trachomatis is the etiological agent of the most common sexually transmitted infection in North America and Europe. Medical complications resulting from genital C. trachomatis infections arise predominantly in women where the initial infections often remain asymptomatic and thus unrecognized. Untreated asymptomatic infections in women can ascend into the upper genital tract and establish persistence, ultimately resulting in extensive scarring of the reproductive organs, pelvic inflammatory disease, infertility and ectopic pregnancies. Previously resolved C. trachomatis infections fail to provide protective immune memory, and no effective vaccine against C. trachomatis is currently available. Critical determinants of the pathogenesis and immunogenicity of genital C. trachomatis infections are cell-autonomous immune responses. Cell-autonomous immunity describes the ability of an individual host cell to launch intrinsic immune circuits that execute the detection, containment and elimination of cell-invading pathogens. As an obligate intracellular pathogen C. trachomatis is constantly under attack by cell-intrinsic host defenses. Accordingly, C. trachomatis evolved to subvert and co-opt cell-autonomous immune pathways. This review will provide a critical summary of our current understanding of cell-autonomous immunity to C. trachomatis and its role in shaping host resistance, inflammation and adaptive immunity to genital C. trachomatis infections.
Collapse
Affiliation(s)
- Ryan Finethy
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC, USA
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
26
|
Evani SJ, Dallo SF, Ramasubramanian AK. Biophysical and Biochemical Outcomes of Chlamydia pneumoniae Infection Promotes Pro-atherogenic Matrix Microenvironment. Front Microbiol 2016; 7:1287. [PMID: 27582738 PMCID: PMC4987350 DOI: 10.3389/fmicb.2016.01287] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 08/04/2016] [Indexed: 01/12/2023] Open
Abstract
Multiple studies support the hypothesis that infectious agents may be involved in the pathogenesis of atherosclerosis. Chlamydia pneumoniae is strongly implicated in atherosclerosis, but the precise role has been underestimated and poorly understood due to the complexity of the disease process. In this work, we test the hypothesis that C. pneumoniae-infected macrophages lodged in the subendothelial matrix contribute to atherogenesis through pro-inflammatory factors and by cell-matrix interactions. To test this hypothesis, we used a 3D infection model with freshly isolated PBMC infected with live C. pneumoniae and chlamydial antigens encapsulated in a collagen matrix, and analyzed the inflammatory responses over 7 days. We observed that infection significantly upregulates the secretion of cytokines TNF-α, IL-1β, IL-8, MCP-1, MMP, oxidative stress, transendothelial permeability, and LDL uptake. We also observed that infected macrophages form clusters, and substantially modify the microstructure and mechanical properties of the extracellular matrix to an atherogenic phenotype. Together, our data demonstrates that C. pneumoniae-infection drives a low-grade, sustained inflammation that may predispose in the transformation to atherosclerotic foci.
Collapse
Affiliation(s)
- Shankar J Evani
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio TX, USA
| | - Shatha F Dallo
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio TX, USA
| | - Anand K Ramasubramanian
- Department of Biomedical Engineering, University of Texas at San Antonio, San AntonioTX, USA; South Texas Center for Emerging Infectious Diseases, San AntonioTX, USA
| |
Collapse
|
27
|
Reimer A, Seufert F, Weiwad M, Ebert J, Bzdyl NM, Kahler CM, Sarkar-Tyson M, Holzgrabe U, Rudel T, Kozjak-Pavlovic V. Inhibitors of macrophage infectivity potentiator-like PPIases affect neisserial and chlamydial pathogenicity. Int J Antimicrob Agents 2016; 48:401-8. [PMID: 27516227 DOI: 10.1016/j.ijantimicag.2016.06.020] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 06/16/2016] [Accepted: 06/18/2016] [Indexed: 10/21/2022]
Abstract
The pathogenic bacteria Chlamydia trachomatis, Neisseria gonorrhoeae and Neisseria meningitidis express the surface-exposed macrophage infectivity potentiator (MIP)-like protein, which plays a role in their pathogenicity. MIP exhibits a peptidyl-prolyl isomerase (PPIase) activity that is inhibited by rapamycin and FK506. In this study, pipecolic acid derivatives were tested for their activity against the chlamydial and neisserial MIP. Two MIP inhibitors were identified, PipN3 and PipN4, that affected the developmental cycle of C. trachomatis in HeLa cells. Furthermore, we could show that deletion of neisserial MIP or addition of the two MIP inhibitors affected the survival of N. gonorrhoeae in the presence of neutrophils. Furthermore, both compounds inhibited the adherence, invasion and/or survival of N. meningitidis in epithelial cells. These results confirm the importance of MIP-like proteins in infection and indicate the relevance of pipecolic acid derivatives as antimicrobials against C. trachomatis, N. gonorrhoeae and N. meningitidis.
Collapse
Affiliation(s)
- Anastasija Reimer
- Biocenter, Chair of Microbiology, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Florian Seufert
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Matthias Weiwad
- Department of Enzymology, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Jutta Ebert
- Biocenter, Chair of Microbiology, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Nicole M Bzdyl
- Marshall Center for Infectious Disease Research and Training, School of Pathology and Laboratory Medicine, University of Western Australia, Perth, WA, Australia
| | - Charlene M Kahler
- Marshall Center for Infectious Disease Research and Training, School of Pathology and Laboratory Medicine, University of Western Australia, Perth, WA, Australia
| | - Mitali Sarkar-Tyson
- Marshall Center for Infectious Disease Research and Training, School of Pathology and Laboratory Medicine, University of Western Australia, Perth, WA, Australia; Defence Science and Technology Laboratory, Biomedical Sciences, Porton Down, Salisbury, Wiltshire SP4 0JQ, UK
| | - Ulrike Holzgrabe
- Institute of Pharmacy and Food Chemistry, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Thomas Rudel
- Biocenter, Chair of Microbiology, University of Würzburg, Am Hubland, 97074 Würzburg, Germany
| | - Vera Kozjak-Pavlovic
- Biocenter, Chair of Microbiology, University of Würzburg, Am Hubland, 97074 Würzburg, Germany.
| |
Collapse
|
28
|
Genital Chlamydia trachomatis: understanding the roles of innate and adaptive immunity in vaccine research. Clin Microbiol Rev 2016; 27:346-70. [PMID: 24696438 DOI: 10.1128/cmr.00105-13] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Chlamydia trachomatis is the leading cause of bacterial sexually transmitted disease worldwide, and despite significant advances in chlamydial research, a prophylactic vaccine has yet to be developed. This Gram-negative obligate intracellular bacterium, which often causes asymptomatic infection, may cause pelvic inflammatory disease (PID), ectopic pregnancies, scarring of the fallopian tubes, miscarriage, and infertility when left untreated. In the genital tract, Chlamydia trachomatis infects primarily epithelial cells and requires Th1 immunity for optimal clearance. This review first focuses on the immune cells important in a chlamydial infection. Second, we summarize the research and challenges associated with developing a chlamydial vaccine that elicits a protective Th1-mediated immune response without inducing adverse immunopathologies.
Collapse
|
29
|
Herweg JA, Rudel T. Interaction of Chlamydiae with human macrophages. FEBS J 2015; 283:608-18. [PMID: 26613554 DOI: 10.1111/febs.13609] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 11/13/2015] [Accepted: 11/24/2015] [Indexed: 11/28/2022]
Abstract
The phylum Chlamydiae contains several members that are well-known human pathogens, like Chlamydia trachomatis and C. pneumoniae. Establishing a chronic bacterial infection requires the active evasion of the host immune response. A major arm of the innate immune defence is constituted by macrophages, which fight infections by removing bacteria and triggering an adaptive immune response. However, some pathogenic Chlamydia infect and survive in macrophages at least for a certain period of time. Therefore, macrophages can serve as vehicles for the dissemination of bacterial infections from the primary infection site via the urogenital or respiratory tract to distant sites in the body. The capacity to infect macrophages seems to depend on the chlamydial strain and the source of macrophages. In vitro infections of macrophages with C. trachomatis, C. psittaci and C. pneumoniae reveal low efficiency of infection and progeny formation, as well as failure to develop mature inclusions. In contrast, the emerging pathogen, Simkania negevensis, actively replicates in macrophages. Here we summarize the current knowledge of the intracellular and molecular key mechanisms of C. trachomatis, C. pneumoniae and S. negevensis infections in human macrophages.
Collapse
Affiliation(s)
- Jo-Ana Herweg
- Biocenter, Department of Microbiology, University of Würzburg, Germany
| | - Thomas Rudel
- Biocenter, Department of Microbiology, University of Würzburg, Germany
| |
Collapse
|
30
|
|
31
|
Humbert MV, Almonacid Mendoza HL, Jackson AC, Hung MC, Bielecka MK, Heckels JE, Christodoulides M. Vaccine potential of bacterial macrophage infectivity potentiator (MIP)-like peptidyl prolyl cis/trans isomerase (PPIase) proteins. Expert Rev Vaccines 2015; 14:1633-49. [PMID: 26468663 DOI: 10.1586/14760584.2015.1095638] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Peptidyl prolyl cis/trans isomerases (PPIases) are a superfamily of proteins ubiquitously distributed among living organisms, which function primarily to assist the folding and structuring of unfolded and partially folded polypeptide chains and proteins. In this review, we focus specifically on the Macrophage Infectivity Potentiator (MIP)-like PPIases, which are members of the immunophilin family of FK506-binding proteins (FKBP). MIP-like PPIases have accessory roles in virulence and are candidates for inclusion in vaccines protective against both animal and human bacterial pathogens. A structural vaccinology approach obviates any issues over molecular mimicry and potential cross-reactivity with human FKBP proteins and studies with a representative antigen, the Neisseria meningitidis-MIP, support this strategy. Moreover, a dual approach of vaccination and drug targeting could be considered for controlling bacterial infectious diseases of humans and animals.
Collapse
Affiliation(s)
- María Victoria Humbert
- a Neisseria Research, Molecular Microbiology, Academic Unit of Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories , University of Southampton Faculty of Medicine , Southampton , UK
| | - Hannia L Almonacid Mendoza
- a Neisseria Research, Molecular Microbiology, Academic Unit of Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories , University of Southampton Faculty of Medicine , Southampton , UK
| | - Alexandra C Jackson
- a Neisseria Research, Molecular Microbiology, Academic Unit of Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories , University of Southampton Faculty of Medicine , Southampton , UK
| | - Miao-Chiu Hung
- a Neisseria Research, Molecular Microbiology, Academic Unit of Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories , University of Southampton Faculty of Medicine , Southampton , UK
| | - Magdalena K Bielecka
- a Neisseria Research, Molecular Microbiology, Academic Unit of Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories , University of Southampton Faculty of Medicine , Southampton , UK
| | - John E Heckels
- a Neisseria Research, Molecular Microbiology, Academic Unit of Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories , University of Southampton Faculty of Medicine , Southampton , UK
| | - Myron Christodoulides
- a Neisseria Research, Molecular Microbiology, Academic Unit of Clinical and Experimental Sciences, Sir Henry Wellcome Laboratories , University of Southampton Faculty of Medicine , Southampton , UK
| |
Collapse
|
32
|
Chlamydia muridarum infection of macrophages elicits bactericidal nitric oxide production via reactive oxygen species and cathepsin B. Infect Immun 2015; 83:3164-75. [PMID: 26015483 DOI: 10.1128/iai.00382-15] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 05/20/2015] [Indexed: 01/13/2023] Open
Abstract
The ability of certain species of Chlamydia to inhibit the biogenesis of phagolysosomes permits their survival and replication within macrophages. The survival of macrophage-adapted chlamydiae correlates with the multiplicity of infection (MOI), and optimal chlamydial growth occurs in macrophages infected at an MOI of ≤1. In this study, we examined the replicative capacity of Chlamydia muridarum in the RAW 264.7 murine macrophage cell line at different MOIs. C. muridarum productively infected these macrophages at low MOIs but yielded few viable elementary bodies (EBs) when macrophages were infected at a moderate (10) or high (100) MOI. While high MOIs caused cytotoxicity and irreversible host cell death, macrophages infected at a moderate MOI did not show signs of cytotoxicity until late in the infectious cycle. Inhibition of host protein synthesis rescued C. muridarum in macrophages infected at a moderate MOI, implying that chlamydial growth was blocked by activated defense mechanisms. Conditioned medium from these macrophages was antichlamydial and contained elevated levels of interleukin 1β (IL-1β), IL-6, IL-10, and beta interferon (IFN-β). Macrophage activation depended on Toll-like receptor 2 (TLR2) signaling, and cytokine production required live, transcriptionally active chlamydiae. A hydroxyl radical scavenger and inhibitors of inducible nitric oxide synthase (iNOS) and cathepsin B also reversed chlamydial killing. High levels of reactive oxygen species (ROS) led to an increase in cathepsin B activity, and pharmacological inhibition of ROS and cathepsin B reduced iNOS expression. Our data demonstrate that MOI-dependent TLR2 activation of macrophages results in iNOS induction via a novel ROS- and cathepsin-dependent mechanism to facilitate C. muridarum clearance.
Collapse
|
33
|
Santiago AE, Mann BJ, Qin A, Cunningham AL, Cole LE, Grassel C, Vogel SN, Levine MM, Barry EM. Characterization of Francisella tularensis Schu S4 defined mutants as live-attenuated vaccine candidates. Pathog Dis 2015; 73:ftv036. [PMID: 25986219 PMCID: PMC4462183 DOI: 10.1093/femspd/ftv036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2015] [Indexed: 01/11/2023] Open
Abstract
Francisella tularensis (Ft), the etiological agent of tularemia and a Tier 1 select agent, has been previously weaponized and remains a high priority for vaccine development. Ft tularensis (type A) and Ft holarctica (type B) cause most human disease. We selected six attenuating genes from the live vaccine strain (LVS; type B), F. novicida and other intracellular bacteria: FTT0507, FTT0584, FTT0742, FTT1019c (guaA), FTT1043 (mip) and FTT1317c (guaB) and created unmarked deletion mutants of each in the highly human virulent Ft strain Schu S4 (Type A) background. FTT0507, FTT0584, FTT0742 and FTT1043 Schu S4 mutants were not attenuated for virulence in vitro or in vivo. In contrast, Schu S4 gua mutants were unable to replicate in murine macrophages and were attenuated in vivo, with an i.n. LD50 > 105 CFU in C57BL/6 mice. However, the gua mutants failed to protect mice against lethal challenge with WT Schu S4, despite demonstrating partial protection in rabbits in a previous study. These results contrast with the highly protective capacity of LVS gua mutants against a lethal LVS challenge in mice, and underscore differences between these strains and the animal models in which they are evaluated, and therefore have important implications for vaccine development. Mutations in guanine biosynthesis genes, but not in four other hypothetical virulence factors in highly virulent Francisella tularensis strain Schu S4 resulted in attenuation in macrophage replication and mouse virulence.
Collapse
Affiliation(s)
- Araceli E Santiago
- Departments of Pediatrics, University of Virginia Children's Hospital, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Barbara J Mann
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Aiping Qin
- Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Aimee L Cunningham
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Leah E Cole
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Christen Grassel
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Myron M Levine
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Eileen M Barry
- Center for Vaccine Development, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
34
|
Co-upregulation of Toll-like receptors 2 and 6 on peripheral blood cells in patients with obstructive sleep apnea. Sleep Breath 2015; 19:873-82. [PMID: 25604476 DOI: 10.1007/s11325-014-1116-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Revised: 12/01/2014] [Accepted: 12/29/2014] [Indexed: 01/01/2023]
Abstract
PURPOSE Toll-like receptor (TLR) 2 can heterodimerise with TLR6 to detect diacylated lipoproteins. Hypoxia inducible factor-1 α co-ordinates selective induction of TLR2 and TLR6 during persistent hypoxia. We hypothesized that TLR 2/6 co-expression may be upregulated by chronic intermittent hypoxia with re-oxygenation (IHR) in obstructive sleep apnea (OSA). METHODS TLR2/6 expressions on blood immune cells were measured in 144 patients with sleep-disordered breathing (SDB), including primary snoring (PS, n = 24), moderate to severe OSA (MSO, n = 60), very severe OSA (VSO, n = 36), and very severe OSA on continuous positive airway pressure (CPAP) treatment (VSOC, n = 24). An in vitro IHR experiment was also undertaken. RESULTS Patients in both the MSO and VSO groups had increased TLR2/6 co-expression on CD16(+) neutrophil than those in the PS group. Patients in the VSOC group had reduced TLR2/6 co-expression on neutrophil than those in either the MSO or VSO group. Blood absolute neutrophil count was positively but weakly correlated with TLR2/6 co-expression on neutrophil. TLR2/6 co-expressions on both CD14(+) monocyte and CD3(+)CD4(+)T helper cell, and TLR2 expressions on both monocyte and T helper cell in SDB patients with low Minimum SaO₂ (≦70%) were all higher than those with high Minimum SaO₂. In vitro IHR for 1-4 days resulted in TLR2/6 co-upregulation on both neutrophil and monocyte. CONCLUSIONS OSA patients had increased TLR2/6 co-expressions on blood immune cells, which were related to their immune cell counts and could be reversed with CPAP treatment. In vitro IHR could induce TLR2/6 co-upregulation.
Collapse
|
35
|
Loffredo S, Staiano RI, Granata F, Costantino V, Borriello F, Frattini A, Lepore MT, Mangoni A, Marone G, Triggiani M. Simplexide induces CD1d-dependent cytokine and chemokine production from human monocytes. PLoS One 2014; 9:e111326. [PMID: 25390653 PMCID: PMC4229102 DOI: 10.1371/journal.pone.0111326] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 09/29/2014] [Indexed: 12/24/2022] Open
Abstract
Monocytes are major effector cells of innate immunity and recognize several endogenous and exogenous molecules due to the expression of wide spectrum of receptors. Among them, the MHC class I-like molecule CD1d interacts with glycolipids and presents them to iNKT cells, mediating their activation. Simplexide belongs to a novel class of glycolipids isolated from marine sponges and is structurally distinct from other immunologically active glycolipids. In this study we have examined the effects of simplexide on cytokine and chemokine release from human monocytes. Simplexide induces a concentration- and time-dependent release of IL-6, CXCL8, TNF-α and IL-10 and increases the expression of IL6, CXCL8 and IL10 mRNA. Cytokine and chemokine release induced by simplexide from monocytes is dependent on CD1d since: i) a CD1d antagonist, 1,2-bis (diphenylphosphino) ethane [DPPE]-polyethylene glycolmonomethylether [PEG], specifically blocks simplexide-induced activation of monocytes; ii) CD1d knockdown inhibits monocyte activation by simplexide and iii) simplexide induces cytokine production from CD1d-transfected but not parental C1R cell line. Finally, we have shown that simplexide also induces iNKT cell expansion in vitro. Our results demonstrate that simplexide, apart from activating iNKT cells, induces the production of cytokines and chemokines from human monocytes by direct interaction with CD1d.
Collapse
Affiliation(s)
- Stefania Loffredo
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Rosaria I. Staiano
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Francescopaolo Granata
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Valeria Costantino
- The NeaNAT group - Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Francesco Borriello
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Annunziata Frattini
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Maria Teresa Lepore
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
| | - Alfonso Mangoni
- The NeaNAT group - Department of Pharmacy, University of Naples Federico II, Naples, Italy
| | - Gianni Marone
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- * E-mail:
| | - Massimo Triggiani
- Division of Allergy and Clinical Immunology, University of Salerno, Salerno, Italy
| |
Collapse
|
36
|
Redgrove KA, McLaughlin EA. The Role of the Immune Response in Chlamydia trachomatis Infection of the Male Genital Tract: A Double-Edged Sword. Front Immunol 2014; 5:534. [PMID: 25386180 PMCID: PMC4209867 DOI: 10.3389/fimmu.2014.00534] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/09/2014] [Indexed: 01/16/2023] Open
Abstract
Chlamydia trachomatis (CT) is the most prevalent bacterial sexually transmitted infection in the world, with more than 100 million cases reported annually. While there have been extensive studies into the adverse effects that CT infection has on the female genital tract, and on the subsequent ability of these women to conceive, studies into the consequences on male fertility have been limited and controversial. This is in part due to the asymptomatic nature of the infection, where it is estimated that 50% of men with Chlamydia fail to show any symptoms. It is accepted, however, that acute and/or persistent CT infection is the causative agent for conditions such as urethritis, epididymitis, epididymo-orchitis, and potentially prostatitis. As with most infections, the immune system plays a fundamental role in the body’s attempts to eradicate the infection. The first and most important immune response to Chlamydia infection is a local one, whereby immune cells such as leukocytes are recruited to the site of infections, and subsequently secrete pro-inflammatory cytokines and chemokines such as interferon gamma. Immune cells also work to initiate and potentiate chronic inflammation through the production of reactive oxygen species (ROS), and the release of molecules with degradative properties including defensins, elastase, collagenase, cathespins, and lysozyme. This long-term inflammation can lead to cell proliferation (a possible precursor to cancer), tissue remodeling, and scarring, as well as being linked to the onset of autoimmune responses in genetically disposed individuals. This review will focus on the ability of the immune system to recognize and clear acute and persistent chlamydial infections in the male genital tract, and on the paradoxical damage that chronic inflammation resulting from the infection can cause on the reproductive health of the individual.
Collapse
Affiliation(s)
- Kate A Redgrove
- Priority Research Centre in Reproductive Biology and Chemical Biology, University of Newcastle , Callaghan, NSW , Australia ; School of Environmental and Life Science, University of Newcastle , Callaghan, NSW , Australia
| | - Eileen A McLaughlin
- Priority Research Centre in Reproductive Biology and Chemical Biology, University of Newcastle , Callaghan, NSW , Australia ; School of Environmental and Life Science, University of Newcastle , Callaghan, NSW , Australia
| |
Collapse
|
37
|
Cheng C, Pal S, Tifrea D, Jia Z, de la Maza LM. A vaccine formulated with a combination of TLR-2 and TLR-9 adjuvants and the recombinant major outer membrane protein elicits a robust immune response and significant protection against a Chlamydia muridarum challenge. Microbes Infect 2014; 16:244-52. [PMID: 24291713 PMCID: PMC3965591 DOI: 10.1016/j.micinf.2013.11.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 11/15/2013] [Accepted: 11/18/2013] [Indexed: 12/17/2022]
Abstract
Chlamydia trachomatis is the most common sexually transmitted bacterial pathogen in the World and there is a need for a vaccine. To enhance the immunogenicity of a vaccine formulated with the Chlamydia muridarum (Cm) mouse pneumonitis recombinant major outer membrane protein (MOMP), we used combinations of Pam2CSK4 + CpG-1826 and Montanide ISA 720 VG + CpG-1826 as adjuvants. Neisseria gonorrhoeae recombinant porin B (Ng-PorB) was used as the antigen control with the same adjuvants. Female BALB/c mice were immunized twice in the nares (i.n.) or in the colon (cl.) and were boosted twice by the intramuscular plus subcutaneous (i.m. + s.c.) routes. Based on the IgG2a/IgG1 ratio in sera, mice immunized with MOMP + Pam2CSK4 + CpG-1826 showed a strong Th2 response while animals vaccinated with MOMP + Montanide ISA 720 VG + CpG-1826 had a Th1 response. Both groups of mice also developed robust Cm-specific T cell proliferation and high levels of IFN-γ. Four weeks after the last immunization, the mice were challenged i.n. with 10(4) inclusion-forming units (IFU) of Cm. Using changes in body weight and number of IFU recovered from the lungs at 10 days post-challenge mice immunized i.n. + i.m./s.c. with MOMP + Pam2CSK4 + CpG-1826 were better protected than other groups. In conclusion, MOMP adjuvanted with Pam2CSK4 + CpG-1826, elicits strong humoral and cellular immune responses and induces significant protection against Chlamydia.
Collapse
Affiliation(s)
- Chunmei Cheng
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA
| | - Sukumar Pal
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA
| | - Delia Tifrea
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA
| | - Zhenyu Jia
- Translational Cancer Biology, Department of Pathology and Laboratory Medicine, University of California, Irvine, Irvine, CA 92697-4800, USA
| | - Luis M de la Maza
- Department of Pathology and Laboratory Medicine, Medical Sciences I, Room D440, University of California, Irvine, Irvine, CA 92697-4800, USA.
| |
Collapse
|
38
|
Signaling via tumor necrosis factor receptor 1 but not Toll-like receptor 2 contributes significantly to hydrosalpinx development following Chlamydia muridarum infection. Infect Immun 2014; 82:1833-9. [PMID: 24549331 DOI: 10.1128/iai.01668-13] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Chlamydial infection in the lower genital tract can lead to hydrosalpinx, which is accompanied by activation of both pattern recognition receptor TLR2- and inflammatory cytokine receptor TNFR1-mediated signaling pathways. In the current study, we compared the relative contributions of these two receptors to chlamydial induction of hydrosalpinx in mice. We found that mice with or without deficiencies in TLR2 or TNFR1 displayed similar time courses of live organism shedding from vaginal swabs, suggesting that these receptor-mediated signaling pathways are not required for controlling chlamydial lower genital infection. However, mice deficient in TNFR1 but not TLR2 developed significantly reduced hydrosalpinx. The decreased pathogenicity correlated with a significant reduction in interleukin-17 by in vitro-restimulated splenocytes of TNFR1-deficient mice. Although TLR2-deficient mice developed hydrosalpinx as severe as that of wild-type mice, peritoneal macrophages from mice deficient in TLR2 but not TNFR1 produced significantly reduced cytokines upon chlamydial stimulation, suggesting that reduced macrophage responses to chlamydial infection do not always lead to a reduction in hydrosalpinx. Thus, we have demonstrated that the signaling pathways triggered by the cytokine receptor TNFR1 play a more significant role in chlamydial induction of hydrosalpinx than those mediated by the pattern recognition receptor TLR2, which has laid a foundation for further revealing the chlamydial pathogenic mechanisms.
Collapse
|
39
|
He J, Wang S, Zeng Y, You X, Ma X, Wu N, Wu Y. Binding of CD14 to Mycoplasma genitalium-Derived Lipid-Associated Membrane Proteins Upregulates TNF-α. Inflammation 2013; 37:322-30. [DOI: 10.1007/s10753-013-9743-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
40
|
Mackern-Oberti JP, Motrich RD, Breser ML, Sánchez LR, Cuffini C, Rivero VE. Chlamydia trachomatis infection of the male genital tract: an update. J Reprod Immunol 2013; 100:37-53. [PMID: 23870458 DOI: 10.1016/j.jri.2013.05.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Revised: 04/03/2013] [Accepted: 05/06/2013] [Indexed: 12/30/2022]
Abstract
Chlamydia trachomatis (CT) is the most prevalent cause of sexually transmitted diseases. Although the prevalence of chlamydial infection is similar in men and women, current research and screening are still focused on women, who develop the most severe complications, leaving the study of male genital tract (MGT) infection underrated. Herein, we reviewed the literature on genital CT infection with special focus on the MGT. Data indicate that CT certainly infects different parts of the MGT such as the urethra, seminal vesicles, prostate, epididymis and testis. However, whether or not CT infection has detrimental effects on male fertility is still controversial. The most important features of CT infection are its chronic nature and the presence of a mild inflammation that remains subclinical in most individuals. Chlamydia antigens and pathogen recognition receptors (PRR), expressed on epithelial cells and immune cells from the MGT, have been studied in the last years. Toll-like receptor (TLR) expression has been observed in the testis, epididymis, prostate and vas deferens. It has been demonstrated that recognition of chlamydial antigens is associated with TLR2, TLR4, and possibly, other PRRs. CT recognition by PRRs induces a local production of cytokines/chemokines, which, in turn, provoke chronic inflammation that might evolve in the onset of an autoimmune process in genetically susceptible individuals. Understanding local immune response along the MGT, as well as the crosstalk between resident leukocytes, epithelial, and stromal cells, would be crucial in inducing a protective immunity, thus adding to the design of new therapeutic approaches to a Chlamydia vaccine.
Collapse
|
41
|
Flavonoid naringenin: a potential immunomodulator for Chlamydia trachomatis inflammation. Mediators Inflamm 2013; 2013:102457. [PMID: 23766556 PMCID: PMC3676976 DOI: 10.1155/2013/102457] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Revised: 04/07/2013] [Accepted: 04/08/2013] [Indexed: 12/21/2022] Open
Abstract
Chlamydia trachomatis, the agent of bacterial sexually transmitted infections, can manifest itself as either acute cervicitis, pelvic inflammatory disease, or a chronic asymptomatic infection. Inflammation induced by C. trachomatis contributes greatly to the pathogenesis of disease. Here we evaluated the anti-inflammatory capacity of naringenin, a polyphenolic compound, to modulate inflammatory mediators produced by mouse J774 macrophages infected with live C. trachomatis. Infected macrophages produced a broad spectrum of inflammatory cytokines (GM-CSF, TNF, IL-1β, IL-1α, IL-6, IL-12p70, and IL-10) and chemokines (CCL4, CCL5, CXCL1, CXCL5, and CXCL10) which were downregulated by naringenin in a dose-dependent manner. Enhanced protein and mRNA gene transcript expressions of TLR2 and TLR4 in addition to the CD86 costimulatory molecule on infected macrophages were modulated by naringenin. Pathway-specific inhibition studies disclosed that p38 mitogen-activated-protein kinase (MAPK) is involved in the production of inflammatory mediators by infected macrophages. Notably, naringenin inhibited the ability of C. trachomatis to phosphorylate p38 in macrophages, suggesting a potential mechanism of its attenuation of concomitantly produced inflammatory mediators. Our data demonstrates that naringenin is an immunomodulator of inflammation triggered by C. trachomatis, which possibly may be mediated upstream by modulation of TLR2, TLR4, and CD86 receptors on infected macrophages and downstream via the p38 MAPK pathway.
Collapse
|
42
|
Kavathas PB, Boeras CM, Mulla MJ, Abrahams VM. Nod1, but not the ASC inflammasome, contributes to induction of IL-1β secretion in human trophoblasts after sensing of Chlamydia trachomatis. Mucosal Immunol 2013; 6:235-43. [PMID: 22763410 PMCID: PMC3465624 DOI: 10.1038/mi.2012.63] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Chlamydia trachomatis (Ct) is an obligate intracellular bacterial pathogen. Previously, we showed that infection of human trophoblast cells by Ct triggers the secretion of the pro-inflammatory cytokine, interleukin (IL)-1β. The aim of this study was to understand the innate immune pathways involved in trophoblast production of IL-1β after Ct infection. The approach we took was to inhibit the expression or function of the key Toll-like receptors (TLRs), Nod-like receptors, and inflammasome components that have been associated with chlamydia infection. In this study, we report that Ct-induced trophoblast IL-1β secretion is associated with the transcription of IL-1β mRNA, the translation and processing of pro-IL-1β, and the activation of caspase-1. In addition, we demonstrate that Ct-induced IL-1β production and secretion by the trophoblast is independent of TLR2, TLR4, MyD88, and the Nalp3/ASC inflammasome. Instead we report, for the first time, the importance of Nod1 for mediating trophoblast IL-1β secretion in response to a Ct infection.
Collapse
Affiliation(s)
- Paula B. Kavathas
- Department of Laboratory Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA,Departments of Immunobiology and Genetics, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA
| | - Crina M. Boeras
- Department of Laboratory Medicine, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA
| | - Melissa J. Mulla
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA
| | - Vikki M. Abrahams
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, 333 Cedar Street, New Haven, CT, USA
| |
Collapse
|
43
|
Toll-like receptor 2-dependent activity of native major outer membrane protein proteosomes of Chlamydia trachomatis. Infect Immun 2012; 81:303-10. [PMID: 23132491 DOI: 10.1128/iai.01062-12] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Chlamydia trachomatis is the most common sexually transmitted bacterial pathogen and the etiologic agent of blinding trachoma. Intracellular signaling pathways leading to host cell inflammation and innate immunity to Chlamydia include those mediated by Toll-like receptors (TLRs) and nucleotide binding oligomerization domain 1 (Nod1) protein. In epithelial cells, TLR-dependent signaling contributes to local immune responses via induction of inflammatory mediators. There is evidence that TLR3, TLR4, and, particularly, TLR2 are critical for Chlamydia-mediated host cell activation and pathology. Despite the importance of TLR2, major chlamydial TLR2 antigens have not been identified so far. Numerous bacterial porins are known TLR2 agonists, i.e., porins from Neisseriae, Shigella, Salmonella, Haemophilus influenzae, and Fusobacterium nucleatum, which share structural and functional similarities with the chlamydial major outer membrane protein (MOMP), a strong antigen candidate for a potential vaccine against C. trachomatis. We describe the ability of purified, detergent-free MOMP to signal via TLR2 in vitro in TLR-overexpressing cells and TLR2-competent human reproductive tract epithelial cell lines. Using MOMP formed in pure protein micelles (proteosomes), we show the induction of TLR2-dependent interleukin-8 (IL-8) and IL-6 secretion in vitro, the involvement of TLR1 as a TLR2 coreceptor, and the activation of both NF-κB and mitogen-activated protein (MAP) kinase intracellular pathways. Interestingly, MOMP proteosomes induce cytokine secretion in endocervical epithelial cells (End/E6E7) but not in urethral epithelial cells (THUECs). A detailed understanding of the TLR2-dependent molecular mechanisms that characterize the effect of MOMP proteosomes on host cells may provide new insights for its successful development as an immunotherapeutic target against Chlamydia.
Collapse
|
44
|
Distinct intensity of host-pathogen interactions in Chlamydia psittaci- and Chlamydia abortus-infected chicken embryos. Infect Immun 2012; 80:2976-88. [PMID: 22689815 DOI: 10.1128/iai.00437-12] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Factors and mechanisms determining the differences in virulence and host specificity between the zoonotic agents Chlamydia psittaci and Chlamydia abortus are still largely unknown. In the present study, two strains were compared for their invasiveness, virulence, and capability of eliciting an immune response in chicken embryos. On breeding day 10, embryonated chicken eggs were inoculated with 5 × 10(4) inclusion-forming units. As shown by immunohistochemistry and quantitative real-time PCR, C. psittaci displayed a significantly better capability of disseminating in the chorioallantoic membrane (CAM) and internal organs than C. abortus. The higher infectious potential of C. psittaci in birds was underlined by significantly higher mRNA expression rates of essential chlamydial genes, such as incA, groEL (in CAM, liver, and spleen), cpaf, and ftsW (in CAM). Although the immune responses to both pathogens were similar, C. psittaci elicited higher macrophage numbers and a stronger expression of a subset of immune-related proteins. The data imply that invasiveness of Chlamydia spp. and propagation in the host are not solely dependent on the level of host immune response but, even to a greater extent, on the expression of bacterial factors related to virulence. The fact that C. psittaci has coped far better than C. abortus with the avian embryo's response by upregulating essential genes may be a key to understanding the mechanisms underlying host adaptation and etiopathology.
Collapse
|
45
|
Siam EM, Hefzy EM. The relationship between antisperm antibodies prevalence and genital Chlamydia trachomatis infection in women with unexplained infertility. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2012. [DOI: 10.1016/j.mefs.2011.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022] Open
|
46
|
Dunn-Siegrist I, Tissières P, Drifte G, Bauer J, Moutel S, Pugin J. Toll-like receptor activation of human cells by synthetic triacylated lipid A-like molecules. J Biol Chem 2012; 287:16121-31. [PMID: 22433865 DOI: 10.1074/jbc.m112.348383] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Recognition of microbial molecules by mammalian host receptors is essential to mount an immune response. Hexaacylated LPS is the prototypic example of a bacterial molecule recognized by the receptor complex TLR4/MD-2 with its lipid A moiety, whereas bacterial lipopeptides are recognized by TLR2. Here we show that a series of synthetic triacylated lipid A-like molecules are weak Toll-like receptor (TLR) agonists (mainly TLR2 agonists) but very potent TLR4/MD-2 antagonists (submicromolar range). Not only do they block human cell responses to LPS but also to whole gram-negative bacteria, and they inhibit the phagocytosis of gram-negative bacteria. These compounds may represent promising immunomodulatory agents.
Collapse
Affiliation(s)
- Irène Dunn-Siegrist
- Intensive Care Laboratory and Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1211 Geneva 14, Switzerland
| | | | | | | | | | | |
Collapse
|
47
|
Rubino I, Coste A, Le Roy D, Roger T, Jaton K, Boeckh M, Monod M, Latgé JP, Calandra T, Bochud PY. Species-specific recognition of Aspergillus fumigatus by Toll-like receptor 1 and Toll-like receptor 6. J Infect Dis 2012; 205:944-54. [PMID: 22315281 DOI: 10.1093/infdis/jir882] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Aspergillus fumigatus causes invasive aspergillosis, a potentially fatal infection in oncohematological patients. Innate immune detection of A. fumigatus involves Toll-like receptor (TLR) 4 and TLR2, which forms a heterodimer with either TLR1 or TLR6. The role of those coreceptors in Aspergillus sensing is unknown. METHODS Cytokine production was measured in bone marrow-derived macrophages (BMDMs) from wild-type (WT) and TLR-deficient mice after incubation with a WT and an immunogenic RodA-deficient (ΔrodA-47) strain of A. fumigatus and in lungs from these mice after intranasal mold inoculation. Aspergillus fumigatus-mediated NF-κB activation was measured in HEK293T cells transfected with plasmids expressing mouse or human TLRs. RESULTS Bone marrow-derived macrophages from TLR1- and TLR6-deficient mice produced lower amounts of interleukin 12p40, CXCL2, interleukin 6, and tumor necrosis factor α than BMDMs from WT mice after stimulation with A. fumigatus. Lungs from TLR1- and TLR6-deficient mice had diminished CXCL1 and CXCL2 production and increased fungal burden after intranasal inoculation of ΔrodA A. fumigatus compared with lungs from WT mice. ΔrodA strain-mediated NF-κB activation was observed in HEK293T cells expressing mouse TLR2/1, mouse TLR2/6, and human TLR2/1 but not human TLR2/6. CONCLUSIONS Innate immune detection of A. fumigatus is mediated by TLR4 and TLR2 together with TLR1 or TLR6 in mice and TLR1 but not TLR6 in humans.
Collapse
Affiliation(s)
- Ivana Rubino
- Department of Medicine, University of Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Coleman CM, Plant K, Newton S, Hobson L, Whyte MKB, Everard ML. The Anti-Apoptotic Effect of Respiratory Syncytial Virus on Human Peripheral Blood Neutrophils is Mediated by a Monocyte Derived Soluble Factor. Open Virol J 2011; 5:114-23. [PMID: 22046209 PMCID: PMC3204419 DOI: 10.2174/1874357901105010114] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Revised: 07/07/2011] [Accepted: 07/27/2011] [Indexed: 12/24/2022] Open
Abstract
Respiratory Syncytial Virus (RSV) causes annual epidemics of respiratory disease particularly affecting infants. The associated airway inflammation is characterized by an intense neutrophilia. This neutrophilic inflammation appears to be responsible for much of the pathology and symptoms. Previous work from our group had shown that there are factors within the airways of infants with RSV bronchiolitis that inhibit neutrophil apoptosis. This study was undertaken to determine if RSV can directly affect neutrophil survival. Neutrophils were isolated from citrated venous blood (collected from healthy adult volunteers) by discontinuous plasma: Percoll gradient centrifugation and, in some experiments, further purified by negative immunomagnetic bead selection. The effect of RSV on neutrophil survival was measured by Annexin V-PE /To-Pro-3 staining and by morphological changes, using Dif-Quick staining of cytospins. Inhibition of neutrophil apoptosis was observed in neutrophils isolated by standard plasma:Percoll gradient when exposed to RSV but not in ultra pure neutrophil preparations. Adding monocytes back to ultra purified preparations restored the effect. The inhibition of apoptosis was observed with both active and UV inactivated virus. The effect is dependent on a soluble factor and appears to be dependent on CD14 receptors on the monocytes.
Collapse
Affiliation(s)
- Christopher M Coleman
- Academic Unit of Child Health, Stephenson Wing, Sheffield Children's Hospital, Sheffield, S10 2TH, UK
| | | | | | | | | | | |
Collapse
|
49
|
Miyairi I, Laxton JD, Wang X, Obert CA, Arva Tatireddigari VRR, van Rooijen N, Hatch TP, Byrne GI. Chlamydia psittaci genetic variants differ in virulence by modulation of host immunity. J Infect Dis 2011; 204:654-63. [PMID: 21791668 DOI: 10.1093/infdis/jir333] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Psittacosis is a zoonosis caused by Chlamydia psittaci and is characterized by severe pneumonia and systemic infection. We sought to determine the basis of the 1000-fold difference in lethal dose of 2 C. psittaci 6BC strains in mice. METHODS Genomes of the strains were sequenced. Mice were infected intraperitoneally and the growth kinetics, immune responses, and pathology were compared. RESULTS The 2 strains differed by the presence of a 7.5-kb plasmid in the attenuated strain and 7 nonsynonomous single-nucleotide polymorphisms between the chromosomes, including a serine/threonine protein kinase gene pkn5. The plasmid was cured from the attenuated strain, but it remained nonlethal. Strains did not differ in growth kinetics in vitro or in vivo. Infection with the attenuated strain led to influx of activated macrophages with relatively minor organ damage. In contrast, the virulent strain caused an influx of nonactivated macrophages, neutrophils, and significant end organ damage. Mice infected with the virulent strain survived challenge when coinfected with either the plasmid-positive or plasmid-negative attenuated strain, indicating that an active process elicited by the attenuated strain reduces inflammation and disease. CONCLUSIONS C. psittaci modulates virulence by alteration of host immunity, which is conferred by small differences in the chromosome.
Collapse
Affiliation(s)
- Isao Miyairi
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Petyaev IM, Zigangirova NA, Tsibezov VV, Ross A, Bashmakov YK. Monoclonal antibodies against lipopolysaccharide of Chlamydia trachomatis with cross reactivity to human ApoB. Hybridoma (Larchmt) 2011; 30:131-6. [PMID: 21529285 DOI: 10.1089/hyb.2010.0107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Splenocytes obtained from mice immunized with whole purified elementary bodies of Chlamydia trachomatis were used for hybridoma construction. The resulting clones were screened with ELISA using chlamydial lipopolysaccharide (LPS) and full-length human apolipoprotein B (ApoB). One analyzed clone producing IgG1 (MAb 7B5) showed simultaneous recognition of chlamydial LPS and human ApoB, suggesting the presence of common antigenic epitopes in their structures. MAb 7B5 exhibited agreeable activity in immunoblot analysis conducted using chlamydial extracts or full-length human ApoB as well as in immunofluorescence (IF) detecting typical inclusion bodies of C. trachomatis and C. pneumoniae in the infected eukaryotic host cells. The removal of LPS from chlamydial suspensions with lauroyl sarcosyl led to a complete disappearance of IF associated with the elementary bodies of C. trachomatis. Therefore, immunologic response to chlamydial antigen may be associated with the generation of ApoB-specific antibody. Molecular mimicry and subsequent formation of cross-reactive antibodies might be an essential mechanism explaining the appearance of circulating auto-antibodies against low density lipoproteins (LDL) in patients with atherosclerosis. Moreover, newly generated MAb 7B5 can be a useful tool in the laboratory diagnosis of chlamydial infections.
Collapse
Affiliation(s)
- Ivan M Petyaev
- Cambridge Theranostics Ltd., Babraham Research Campus, Babraham, Cambridge, United Kingdom
| | | | | | | | | |
Collapse
|