1
|
Kumari S, Martinez-Garcia J, Ehrman RN, Tang W, Miles J, Basak P, Howlett TS, Wijesundara YH, Wang Z, Izzo A, Restrepo B, Lu L, Meloni G, Gassensmith JJ. Testing the Antigenic Potential of Transmembrane Proteins To Develop a Thermostable Tuberculosis MOF-Liposomal Vaccine. ACS Infect Dis 2024. [PMID: 39663556 DOI: 10.1021/acsinfecdis.4c00771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Tuberculosis is one of the deadliest infectious diseases and continues to be a major health risk in many parts of the world. Even today, the century-old Bacillus Calmette-Guerin (BCG) vaccine is the only formulation on the market and is ineffective for several sections of the global population responsible for transmission. In the search for antigens that can mount a robust immune response, we have reported the recombinant expression and purification of two novel membrane proteins, the Cation transporter protein V (CtpV) and the Mycobacterial copper transporter B (MctB) present on the membrane surface of Mycobacterium tuberculosis. CtpV was tested as an antigen against the plasma of tuberculosis patients and was found to have a unique immune response profile compared with more commonly studied tuberculosis (TB) antigens. CtpV and MctB were reconstituted into proteoliposomes─individually and in combination─to stabilize them in a lipid bilayer and create a nanoparticle vaccine platform. In vivo experiments demonstrated that when delivered with an adjuvant, these antigens generated a robust Th1-biased T-cell response in mice, with the combination of both antigens performing the best and generating a response comparable to BCG. Since tuberculosis vaccines often need to be shipped to areas with fluctuating power supply, we encapsulated the proteoliposomes and the adjuvant in ZIF-8 to create a shelf-stable formulation. Complementary in vivo studies were carried out to confirm that the ZIF-8 coating did not interfere with or compromise the immunogenicity of the antigens.
Collapse
Affiliation(s)
- Sneha Kumari
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Jonathan Martinez-Garcia
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Ryanne Nicole Ehrman
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Wendy Tang
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Joshua Miles
- Department of Immunology, UT Southwestern Medical Center, Dallas, Texas 75390, United States
| | - Priyanka Basak
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Thomas Sinclair Howlett
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Yalini Hansika Wijesundara
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Ziqi Wang
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Angelo Izzo
- Centenary Institute, University of Sydney, Sydney, NSW2050, Australia
| | - Blanca Restrepo
- School of Public Health at Brownsville, University of Texas Health Science Center at Houston, Brownsville, Texas 78520, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Edinburg, Texas 78539, United States
| | - Lenette Lu
- Department of Immunology, UT Southwestern Medical Center, Dallas, Texas 75390, United States
- Division of Infectious Diseases and Geographic Medicine, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas 75390, United States
- Parkland Health & Hospital System, Dallas, Texas 75235, United States
| | - Gabriele Meloni
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
| | - Jeremiah J Gassensmith
- Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, Texas 75080, United States
- Department of Bioengineering, University of Texas at Dallas, Richardson, Texas 75080, United States
| |
Collapse
|
2
|
Timm MR, Russell SK, Hultgren SJ. Urinary tract infections: pathogenesis, host susceptibility and emerging therapeutics. Nat Rev Microbiol 2024:10.1038/s41579-024-01092-4. [PMID: 39251839 DOI: 10.1038/s41579-024-01092-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/11/2024]
Abstract
Urinary tract infections (UTIs), which include any infection of the urethra, bladder or kidneys, account for an estimated 400 million infections and billions of dollars in health-care spending per year. The most common bacterium implicated in UTI is uropathogenic Escherichia coli, but diverse pathogens including Klebsiella, Enterococcus, Pseudomonas, Staphylococcus and even yeast such as Candida species can also cause UTIs. UTIs occur in both women and men and in both healthy and immunocompromised patients. However, certain patient factors predispose to disease: for example, female sex, history of prior UTI, or the presence of a urinary catheter or other urinary tract abnormality. The current clinical paradigm for the treatment of UTIs involves the use of antibiotics. Unfortunately, the efficacy of this approach is dwindling as the prevalence of antimicrobial resistance rises among UTI isolates, and the immense quantity of antibiotics prescribed annually for these infections contributes to the emergence of resistant pathogens. Therefore, there is an urgent need for new antibiotics and non-antibiotic treatment and prevention strategies. In this Review, we discuss how recent studies of bacterial pathogenesis, recurrence, persistence, host-pathogen interactions and host susceptibility factors have elucidated new and promising targets for the treatment and prevention of UTIs.
Collapse
Affiliation(s)
- Morgan R Timm
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
| | - Seongmi K Russell
- Department of Paediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Scott J Hultgren
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
- Center for Women's Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
3
|
Frick-Cheng AE, Shea AE, Roberts JR, Smith SN, Ohi MD, Mobley HLT. Iron limitation induces motility in uropathogenic E. coli CFT073 partially through action of LpdA. mBio 2024; 15:e0104824. [PMID: 38874412 PMCID: PMC11253704 DOI: 10.1128/mbio.01048-24] [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: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 06/15/2024] Open
Abstract
More than half of women will experience a urinary tract infection (UTI) with most cases caused by uropathogenic Escherichia coli (UPEC). Bacterial swimming motility enhances UPEC pathogenicity, resulting in more severe disease outcomes including kidney infection. Surprisingly, the connection between motility and iron limitation is mostly unexplored despite the lack of free iron available in the host. We sought to investigate a potential connection between iron restriction and regulation of motility in UPEC. We cultured E. coli CFT073, a prototypical UPEC strain, under iron limitation and observed that CFT073 had elevated fliC (flagella) promoter activity, and this iron-specific response was repressed by the addition of exogenous iron. We confirmed increased flagellar expression in CFT073 by measuring fliC transcript, FliC protein, and surface-expressed flagella under iron-limited conditions. Interestingly, known motility regulator flhDC did not have altered transcription under these conditions. To define the regulatory mechanism of this response, we constructed single knockouts of eight master regulators and found the iron-regulated response was lost in crp, arcA, and fis mutants. Thus, we focused on the five genes regulated by all three regulators. Of the five genes knocked out, the iron-regulated motility response was most strongly dysregulated in the lpdA mutant, which also resulted in significantly lowered fitness in the murine model of ascending UTI, both against the WT and a non-motile fliC mutant. Collectively, we demonstrated that iron-mediated motility in CFT073 is partially regulated by lpdA, which contributes to the understanding of how uropathogens differentially regulate motility mechanisms in the iron-restricted host. IMPORTANCE Urinary tract infections (UTIs) are ubiquitous and responsible for over five billion dollars in associated health care costs annually. Both iron acquisition and motility are highly studied virulence factors associated with uropathogenic Escherichia coli (UPEC), the main causative agent of uncomplicated UTI. This work is innovative by providing mechanistic insight into the synergistic relationship between these two critical virulence properties. Here, we demonstrate that iron limitation has pleiotropic effects with consequences that extend beyond metabolism and impact other virulence mechanisms. Indeed, targeting iron acquisition as a therapy may lead to an undesirable enhancement of UPEC pathogenesis through increased motility. It is vital to understand the full breadth of UPEC pathogenesis to adequately respond to this common infection, especially with the increase of antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- A. E. Frick-Cheng
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - A. E. Shea
- Department of Microbiology and Immunology, University of South Alabama Medical School, Mobile, Alabama, USA
| | - J. R. Roberts
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - S. N. Smith
- Department of Anesthesiology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - M. D. Ohi
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - H. L. T. Mobley
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
Saz-Leal P, Ligon MM, Diez-Rivero CM, García-Ayuso D, Mohanty S, Viñuela M, Real-Arévalo I, Conejero L, Brauner A, Subiza JL, Mysorekar IU. MV140 Mucosal Vaccine Induces Targeted Immune Response for Enhanced Clearance of Uropathogenic E. coli in Experimental Urinary Tract Infection. Vaccines (Basel) 2024; 12:535. [PMID: 38793786 PMCID: PMC11126127 DOI: 10.3390/vaccines12050535] [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: 04/02/2024] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
MV140 is an inactivated whole-cell bacterial mucosal vaccine with proven clinical efficacy against recurrent urinary tract infections (UTIs). These infections are primarily caused by uropathogenic E. coli (UPEC) strains, which are unique in their virulence factors and remarkably diverse. MV140 contains a non-UPEC strain, suggesting that it may induce an immune response against different UPEC-induced UTIs in patients. To verify this, we experimentally evaluated the cellular and humoral responses to UTI89, a prototypical UPEC strain, in mice vaccinated with MV140, as well as the degree of protection achieved in a UPEC UTI89 model of acute cystitis. The results show that both cellular (Th1/Th17) and antibody (IgG/IgA) responses to UTI89 were induced in MV140-immunized mice. MV140 vaccination resulted in an early increased clearance of UTI89 viable bacteria in the bladder and urine following transurethral infection. This was accompanied by a highly significant increase in CD4+ T cells in the bladder and an increase in urinary neutrophils. Collectively, our results support that MV140 induces cross-reactive humoral and cellular immune responses and cross-protection against UPEC strains.
Collapse
Affiliation(s)
- Paula Saz-Leal
- Inmunotek S.L., 28805 Madrid, Spain; (C.M.D.-R.); (D.G.-A.); (M.V.); (I.R.-A.); (L.C.); (J.L.S.)
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA; (M.M.L.); (I.U.M.)
| | - Marianne Morris Ligon
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA; (M.M.L.); (I.U.M.)
| | - Carmen María Diez-Rivero
- Inmunotek S.L., 28805 Madrid, Spain; (C.M.D.-R.); (D.G.-A.); (M.V.); (I.R.-A.); (L.C.); (J.L.S.)
| | - Diego García-Ayuso
- Inmunotek S.L., 28805 Madrid, Spain; (C.M.D.-R.); (D.G.-A.); (M.V.); (I.R.-A.); (L.C.); (J.L.S.)
| | - Soumitra Mohanty
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.M.); (A.B.)
- Division of Clinical Microbiology, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - Marcos Viñuela
- Inmunotek S.L., 28805 Madrid, Spain; (C.M.D.-R.); (D.G.-A.); (M.V.); (I.R.-A.); (L.C.); (J.L.S.)
| | - Irene Real-Arévalo
- Inmunotek S.L., 28805 Madrid, Spain; (C.M.D.-R.); (D.G.-A.); (M.V.); (I.R.-A.); (L.C.); (J.L.S.)
| | - Laura Conejero
- Inmunotek S.L., 28805 Madrid, Spain; (C.M.D.-R.); (D.G.-A.); (M.V.); (I.R.-A.); (L.C.); (J.L.S.)
| | - Annelie Brauner
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden; (S.M.); (A.B.)
- Division of Clinical Microbiology, Karolinska University Hospital, 17176 Stockholm, Sweden
| | - José Luis Subiza
- Inmunotek S.L., 28805 Madrid, Spain; (C.M.D.-R.); (D.G.-A.); (M.V.); (I.R.-A.); (L.C.); (J.L.S.)
| | - Indira Uppugunduri Mysorekar
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO 63110, USA; (M.M.L.); (I.U.M.)
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Xing Y, Clark JR, Chang JD, Zulk JJ, Chirman DM, Piedra FA, Vaughan EE, Hernandez Santos HJ, Patras KA, Maresso AW. Progress toward a vaccine for extraintestinal pathogenic E. coli (ExPEC) II: efficacy of a toxin-autotransporter dual antigen approach. Infect Immun 2024; 92:e0044023. [PMID: 38591882 PMCID: PMC11075464 DOI: 10.1128/iai.00440-23] [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: 11/01/2023] [Accepted: 03/18/2024] [Indexed: 04/10/2024] Open
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) is a leading cause of worldwide morbidity and mortality, the top cause of antimicrobial-resistant (AMR) infections, and the most frequent cause of life-threatening sepsis and urinary tract infections (UTI) in adults. The development of an effective and universal vaccine is complicated by this pathogen's pan-genome, its ability to mix and match virulence factors and AMR genes via horizontal gene transfer, an inability to decipher commensal from pathogens, and its intimate association and co-evolution with mammals. Using a pan virulome analysis of >20,000 sequenced E. coli strains, we identified the secreted cytolysin α-hemolysin (HlyA) as a high priority target for vaccine exploration studies. We demonstrate that a catalytically inactive pure form of HlyA, expressed in an autologous host using its own secretion system, is highly immunogenic in a murine host, protects against several forms of ExPEC infection (including lethal bacteremia), and significantly lowers bacterial burdens in multiple organ systems. Interestingly, the combination of a previously reported autotransporter (SinH) with HlyA was notably effective, inducing near complete protection against lethal challenge, including commonly used infection strains ST73 (CFT073) and ST95 (UTI89), as well as a mixture of 10 of the most highly virulent sequence types and strains from our clinical collection. Both HlyA and HlyA-SinH combinations also afforded some protection against UTI89 colonization in a murine UTI model. These findings suggest recombinant, inactive hemolysin and/or its combination with SinH warrant investigation in the development of an E. coli vaccine against invasive disease.
Collapse
Affiliation(s)
- Yikun Xing
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Justin R. Clark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - James D. Chang
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Jacob J. Zulk
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Dylan M. Chirman
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Felipe-Andres Piedra
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Ellen E. Vaughan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Haroldo J. Hernandez Santos
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| | - Kathryn A. Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, USA
| | - Anthony W. Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
6
|
Westcott MM, Morse AE, Troy G, Blevins M, Wierzba T, Sanders JW. Photochemical inactivation as an alternative method to produce a whole-cell vaccine for uropathogenic Escherichia coli (UPEC). Microbiol Spectr 2024; 12:e0366123. [PMID: 38315025 PMCID: PMC10913755 DOI: 10.1128/spectrum.03661-23] [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: 10/20/2023] [Accepted: 01/10/2024] [Indexed: 02/07/2024] Open
Abstract
Uropathogenic Escherichia coli (UPEC) is the primary causative agent of lower urinary tract infection (UTI). UTI presents a serious health risk and has considerable secondary implications including economic burden, recurring episodes, and overuse of antibiotics. A safe and effective vaccine would address this widespread health problem and emerging antibiotic resistance. Killed, whole-cell vaccines have shown limited efficacy to prevent recurrent UTI in human trials. We explored photochemical inactivation with psoralen drugs and UVA light (PUVA), which crosslinks nucleic acid, as an alternative to protein-damaging methods of inactivation to improve whole-cell UTI vaccines. Exposure of UPEC to the psoralen drug AMT and UVA light resulted in a killed but metabolically active (KBMA) state, as reported previously for other PUVA-inactivated bacteria. The immunogenicity of PUVA-UPEC as compared to formalin-inactivated UPEC was compared in mice. Both generated high UPEC-specific serum IgG titers after intramuscular delivery. However, using functional adherence as a measure of surface protein integrity, we found differences in the properties of PUVA- and formalin-inactivated UPEC. Adhesion mediated by Type-1 and P-fimbriae was severely compromised by formalin but was unaffected by PUVA, indicating that PUVA preserved the functional conformation of fimbrial proteins, which are targets of protective immune responses. In vitro assays indicated that although they retained metabolic activity, PUVA-UPEC lost virulence properties that could negatively impact vaccine safety. Our results imply the potential for PUVA to improve killed, whole-cell UTI vaccines by generating bacteria that more closely resemble their live, infectious counterparts relative to vaccines generated with protein-damaging methods. IMPORTANCE Lower urinary tract infection (UTI), caused primarily by uropathogenic Escherichia coli, represents a significant health burden, accounting for 7 million primary care and 1 million emergency room visits annually in the United States. Women and the elderly are especially susceptible and recurrent infection (rUTI) is common in those populations. Lower UTI can lead to life-threatening systemic infection. UTI burden is manifested by healthcare dollars spent (1.5 billion annually), quality of life impact, and resistant strains emerging from antibiotic overuse. A safe and effective vaccine to prevent rUTI would address a substantial healthcare issue. Vaccines comprised of inactivated uropathogenic bacteria have yielded encouraging results in clinical trials but improvements that enhance vaccine performance are needed. To that end, we focused on inactivation methodology and provided data to support photochemical inactivation, which targets nucleic acid, as a promising alternative to conventional protein-damaging inactivation methods to improve whole-cell UTI vaccines.
Collapse
Affiliation(s)
- Marlena M. Westcott
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| | - Alexis E. Morse
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| | - Gavin Troy
- Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| | - Maria Blevins
- Department of Internal Medicine, Infectious Diseases Section, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| | - Thomas Wierzba
- Department of Internal Medicine, Infectious Diseases Section, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| | - John W. Sanders
- Department of Internal Medicine, Infectious Diseases Section, Wake Forest University School of Medicine, Winston Salem, North Carolina, USA
| |
Collapse
|
7
|
Gharaibeh MH, Sheyab SYA, Lafi SQ, Etoom EM. Risk factors associated with mcr-1 colistin-resistance gene in Escherichia coli broiler samples in northern Jordan. J Glob Antimicrob Resist 2024; 36:284-292. [PMID: 38325733 DOI: 10.1016/j.jgar.2024.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 01/06/2024] [Accepted: 01/10/2024] [Indexed: 02/09/2024] Open
Abstract
OBJECTIVES The purpose of this study was to determine the prevalence of colistin-resistant Escherichia coli carrying mcr-1, and to identify risk factors associated with mcr gene-mediated resistance. METHODS In total, 385 cloacal samples were collected from 125 broiler farms and a questionnaire containing information about each farm was designed and filled. RESULTS Most of the antibiotics used in the disk diffusion method were highly resistant in all samples, with tetracycline and penicillin showing 100% and 99.7% resistance, respectively. Additionally, avian pathogenic E. coli (APEC) virulence genes frequency and percentage of APEC were identified, including sitA,iucC, and astA at 77%, 70.5%, and 62% respectively. In total, 214 of 360 isolates were positive for APEC (59.4%). Based on the minimum inhibitory (MIC) test, 58% of the isolates (n = 209 of 360) were resistant to colistin, with 39.7% displaying the mcr-1 gene. The statistical analysis of risk factors that influence colistin resistance prevalence revealed several significant factors, including commercial feed, farm management, sanitization, and antibiotic use. Irregular health checks for workers, non-dipping of feet before entering poultry houses, and the use of commercial poultry feeds all contributed to higher levels of colistin resistance as measured by MIC. On the other hand, doxycycline and commercial feed was 4 and 3.2 times more likely to occur based on the final logistic model of the mcr-1 gene, respectively. CONCLUSION Our results suggest that better biosecurity protocols should be implemented in poultry farms to reduce antibiotic-resistant bacteria. Additionally, antibiotics should be carefully monitored and used only when necessary.
Collapse
Affiliation(s)
- Mohammad H Gharaibeh
- Department of Basic Veterinary Medical Science, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid, Jordan.
| | - Sahba Y Al Sheyab
- Department of Basic Veterinary Medical Science, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Shawkat Q Lafi
- Department of Pathology and Public Health, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | - Eman M Etoom
- Department of Basic Veterinary Medical Science, Faculty of Veterinary Medicine, Jordan University of Science and Technology, Irbid, Jordan
| |
Collapse
|
8
|
Qiu L, Chirman D, Clark JR, Xing Y, Hernandez Santos H, Vaughan EE, Maresso AW. Vaccines against extraintestinal pathogenic Escherichia coli (ExPEC): progress and challenges. Gut Microbes 2024; 16:2359691. [PMID: 38825856 PMCID: PMC11152113 DOI: 10.1080/19490976.2024.2359691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 05/21/2024] [Indexed: 06/04/2024] Open
Abstract
The emergence of antimicrobial resistance (AMR) is a principal global health crisis projected to cause 10 million deaths annually worldwide by 2050. While the Gram-negative bacteria Escherichia coli is commonly found as a commensal microbe in the human gut, some strains are dangerously pathogenic, contributing to the highest AMR-associated mortality. Strains of E. coli that can translocate from the gastrointestinal tract to distal sites, called extraintestinal E. coli (ExPEC), are particularly problematic and predominantly afflict women, the elderly, and immunocompromised populations. Despite nearly 40 years of clinical trials, there is still no vaccine against ExPEC. One reason for this is the remarkable diversity in the ExPEC pangenome across pathotypes, clades, and strains, with hundreds of genes associated with pathogenesis including toxins, adhesins, and nutrient acquisition systems. Further, ExPEC is intimately associated with human mucosal surfaces and has evolved creative strategies to avoid the immune system. This review summarizes previous and ongoing preclinical and clinical ExPEC vaccine research efforts to help identify key gaps in knowledge and remaining challenges.
Collapse
Affiliation(s)
- Ling Qiu
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Dylan Chirman
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Justin R. Clark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Tailored Antibacterials and Innovative Laboratories for Phage (Φ) Research (TAILΦR), Baylor College of Medicine, Houston, TX, USA
| | - Yikun Xing
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Haroldo Hernandez Santos
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Tailored Antibacterials and Innovative Laboratories for Phage (Φ) Research (TAILΦR), Baylor College of Medicine, Houston, TX, USA
| | - Ellen E. Vaughan
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Anthony W. Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Tailored Antibacterials and Innovative Laboratories for Phage (Φ) Research (TAILΦR), Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
9
|
Wang Y, Perepelov AV, Senchenkova SN, Lu G, Wang X, Ma G, Yang Q, Yuan J, Wang Y, Xie L, Jiang X, Qin J, Liu D, Liu M, Huang D, Liu B. Glycoengineering directs de novo biomanufacturing of UPEC O21 O-antigen polysaccharide based glycoprotein. Int J Biol Macromol 2023; 253:126993. [PMID: 37739281 DOI: 10.1016/j.ijbiomac.2023.126993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/06/2023] [Accepted: 09/17/2023] [Indexed: 09/24/2023]
Abstract
Glycoproteins, in which polysaccharides are usually attached to proteins, are an important class of biomolecules that are widely used as therapeutic agents in clinical treatments for decades. Uropathogenic Escherichia coli (UPEC) O21 has been identified as a serogroup that induces urinary tract infections, with a global increasing number among women and young children. Therefore, there is an urgent need to establish protective vaccines against UPEC infection. Herein, we engineered non-pathogenic E. coli MG1655 to achieve robust, cost-effective de novo biosynthesis of O21 O-antigen polysaccharide-based glycoprotein against UPEC O21. Specifically, this glycoengineered E. coli MG1655 was manipulated for high-efficient glucose-glycerol co-utilization and for the gene cluster installation and O-glycosylation machinery assembly. The key pathways of UDP-sugar precursors were also strengthened to enforce more carbon flux towards the glycosyl donors, which enhanced the glycoprotein titer by 5.6-fold. Further optimization of culture conditions yielded glycoproteins of up to 35.34 mg/L. Glycopeptide MS confirmed the preciset biosynthesis of glycoprotein. This glycoprotein elicited antigen-specific IgG immune responses and significantly reduced kidney and bladder colonization. This bacterial cell-based glyco-platform and optimized strategies can provide a guideline for the biosynthesis of other value-added glycoproteins.
Collapse
Affiliation(s)
- Yuhui Wang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China; National Glycoengineering Research Center, Shandong University, Qingdao, Shandong 266237, China
| | - Andrei V Perepelov
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 119991 Moscow, Russian Federation
| | - Sof'ya N Senchenkova
- N. D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 119991 Moscow, Russian Federation
| | - Gege Lu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Xiaohan Wang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Guozhen Ma
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Qian Yang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Jian Yuan
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Yanling Wang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Lijie Xie
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Xiaolong Jiang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Jingliang Qin
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Dan Liu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Miaomiao Liu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China
| | - Di Huang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China.
| | - Bin Liu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, Nankai University, Tianjin, China; TEDA Institute of Biological Sciences and Biotechnology, Tianjin Key Laboratory of Microbial Functional Genomics, Nankai University, Tianjin, China.
| |
Collapse
|
10
|
Crothers JW, Norton EB. Recent advances in enterotoxin vaccine adjuvants. Curr Opin Immunol 2023; 85:102398. [PMID: 37976963 PMCID: PMC11258862 DOI: 10.1016/j.coi.2023.102398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/21/2023] [Accepted: 10/18/2023] [Indexed: 11/19/2023]
Abstract
Enterotoxin adjuvants have been researched for their ability to promote immunity to co-delivered antigens. Outside of cholera vaccines, however, these proteins have yet to be included in any currently licensed vaccines. They include molecules derived from the bacterial toxins of Vibrio cholerae, cholera toxin, or Escherichia coli, heat-labile toxin, such as detoxified mutants or subunits. This class of adjuvants is distinguished by their delivery possibilities, which include parenteral injection, skin applications, or direct mucosal administration by oral, sublingual, or nasal routes. In addition, inclusion of an enterotoxin adjuvant is associated with development of multifaceted cellular and humoral immune responses to vaccination. Here, we review exciting progress in the past few years in clinical trials for safety and efficacy, preclinical vaccines studies, and new mechanistic insights for enterotoxin adjuvants. This includes recent reports of their use in vaccines targeting microbial infections (bacterial, viral, parasitic) or substance abuse drugs.
Collapse
Affiliation(s)
- Jessica W Crothers
- Department of Pathology and Laboratory Medicine, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | | |
Collapse
|
11
|
Hawas S, Vagenas D, Haque A, Totsika M. Bladder-draining lymph nodes support germinal center B cell responses during urinary tract infection in mice. Infect Immun 2023; 91:e0031723. [PMID: 37882531 PMCID: PMC10652902 DOI: 10.1128/iai.00317-23] [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: 08/12/2023] [Accepted: 10/02/2023] [Indexed: 10/27/2023] Open
Abstract
Bacterial urinary tract infections (UTIs) are both common and exhibit high recurrence rates in women. UTI healthcare costs are increasing due to the rise of multidrug-resistant (MDR) bacteria, necessitating alternative approaches for infection control. Here, we directly observed host adaptive immune responses in acute UTI. We employed a mouse model in which wild-type C57BL/6J mice were transurethrally inoculated with a clinically relevant MDR UTI strain of uropathogenic Escherichia coli (UPEC). Firstly, we noted that rag1-/- C57BL/6J mice harbored larger bacterial burdens than wild-type counterparts, consistent with a role for adaptive immunity in UTI control. Consistent with this, UTI triggered in the bladders of wild-type mice early increases of myeloid cells, including CD11chi conventional dendritic cells, suggesting possible involvement of these professional antigen-presenting cells. Importantly, germinal center B cell responses developed by 4 weeks post-infection in bladder-draining lymph nodes of wild-type mice and, although modest in magnitude and transient in nature, could not be boosted with a second UTI. Thus, our data reveal for the first time in a mouse model that UPEC UTI induces local B cell immune responses in bladder-draining lymph nodes, which could potentially serve to control infection.
Collapse
Affiliation(s)
- Sophia Hawas
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Dimitrios Vagenas
- Research Methods Group, School of Public Health and Social Work, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Ashraful Haque
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
- Department of Microbiology and Immunology, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia
| | - Makrina Totsika
- Centre for Immunology and Infection Control, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
12
|
Goubet AG, Rouanne M, Derosa L, Kroemer G, Zitvogel L. From mucosal infection to successful cancer immunotherapy. Nat Rev Urol 2023; 20:682-700. [PMID: 37433926 DOI: 10.1038/s41585-023-00784-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2023] [Indexed: 07/13/2023]
Abstract
The clinical management of advanced malignancies of the upper and lower urinary tract has been revolutionized with the advent of immune checkpoint blockers (ICBs). ICBs reinstate or bolster pre-existing immune responses while creating new T cell specificities. Immunogenic cancers, which tend to benefit more from immunotherapy than cold tumours, harbour tumour-specific neoantigens, often associated with a high tumour mutational burden, as well as CD8+ T cell infiltrates and ectopic lymphoid structures. The identification of beneficial non-self tumour antigens and natural adjuvants is the focus of current investigation. Moreover, growing evidence suggests that urinary or intestinal commensals, BCG and uropathogenic Escherichia coli influence long-term responses in patients with kidney or bladder cancer treated with ICBs. Bacteria infecting urothelium could be a prominent target for T follicular helper cells and B cells, linking innate and cognate CD8+ memory responses. In the urinary tract, commensal flora differ between healthy and tumoural mucosae. Although antibiotics can affect the prognosis of urinary tract malignancies, bacteria can have a major influence on cancer immunosurveillance. Beyond their role as biomarkers, immune responses against uropathogenic commensals could be harnessed for the design of future immunoadjuvants that can be advantageously combined with ICBs.
Collapse
Affiliation(s)
- Anne-Gaëlle Goubet
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Department of Pathology and Immunology, University of Geneva, Geneva, Switzerland
- AGORA Cancer Center, Lausanne, Switzerland
| | - Mathieu Rouanne
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Lisa Derosa
- Gustave Roussy, Villejuif, France
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicetre, France
| | - Guido Kroemer
- Gustave Roussy, Villejuif, France
- Equipe labellisée par la Ligue contre le Cancer, Université de Paris Cité, Sorbonne Université, Institut Universitaire de France, Inserm U1138, Centre de Recherche des Cordeliers, Paris, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Laurence Zitvogel
- Gustave Roussy, Villejuif, France.
- Institut National de la Santé Et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée - Ligue Nationale contre le Cancer, Villejuif, France.
- Faculté de Médecine, Université Paris-Saclay, Kremlin-Bicetre, France.
- Center of Clinical Investigations for In Situ Biotherapies of Cancer (BIOTHERIS) INSERM, CIC1428, Villejuif, France.
| |
Collapse
|
13
|
Schwartz L, de Dios Ruiz-Rosado J, Stonebrook E, Becknell B, Spencer JD. Uropathogen and host responses in pyelonephritis. Nat Rev Nephrol 2023; 19:658-671. [PMID: 37479904 PMCID: PMC10913074 DOI: 10.1038/s41581-023-00737-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/23/2023]
Abstract
Urinary tract infections (UTIs) are among the most common bacterial infections seen in clinical practice. The ascent of UTI-causing pathogens to the kidneys results in pyelonephritis, which can trigger kidney injury, scarring and ultimately impair kidney function. Despite sizable efforts to understand how infections develop or are cleared in the bladder, our appreciation of the mechanisms by which infections develop, progress or are eradicated in the kidney is limited. The identification of virulence factors that are produced by uropathogenic Escherichia coli to promote pyelonephritis have begun to fill this knowledge gap, as have insights into the mechanisms by which kidney tubular epithelial cells oppose uropathogenic E. coli infection to prevent or eradicate UTIs. Emerging data also illustrate how specific cellular immune responses eradicate infection whereas other immune cell populations promote kidney injury. Insights into the mechanisms by which uropathogenic E. coli circumvent host immune defences or antibiotic therapy to cause pyelonephritis is paramount to the development of new prevention and treatment strategies to mitigate pyelonephritis and its associated complications.
Collapse
Affiliation(s)
- Laura Schwartz
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA.
- The Ohio State University College of Medicine, Columbus, OH, USA.
| | - Juan de Dios Ruiz-Rosado
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Emily Stonebrook
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Brian Becknell
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - John David Spencer
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA.
- The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
14
|
Frick-Cheng AE, Shea AE, Roberts JR, Smith SN, Ohi MD, Mobley HLT. Altered motility in response to iron-limitation is regulated by lpdA in uropathogenic E. coli CFT073. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559868. [PMID: 37808639 PMCID: PMC10557643 DOI: 10.1101/2023.09.27.559868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
More than half of all women will experience a urinary tract infection (UTI) in their lifetime with most cases caused by uropathogenic Escherichia coli (UPEC). Bacterial motility enhances UPEC pathogenicity, resulting in more severe disease outcomes including kidney infection. Surprisingly, the connection between motility and iron limitation is mostly unexplored, despite the lack of free iron available in the host. Therefore, we sought to explore the potential connection between iron restriction and regulation of motility in UPEC. We cultured E. coli CFT073, a prototypical UPEC strain, in media containing an iron chelator. Under iron limitation, CFT073 had elevated fliC (flagella) promoter activity, driving motility on the leading edge of the colony. Furthermore, this iron-specific response was repressed by the addition of exogenous iron. We confirmed increased flagella expression in CFT073 by measuring fliC transcript, FliC protein, and surface-expressed flagella under iron-limited conditions. To define the regulatory mechanism, we constructed single knockouts of eight master regulators. The iron-regulated response was lost in crp, arcA, and fis mutants. Thus, we focused on the five genes regulated by all three transcription factors. Of the five genes knocked out, the iron-regulated motility response was most strongly dysregulated in an lpdA mutant, which also resulted in significantly lowered fitness in the murine model of ascending UTI. Collectively, we demonstrated that iron-mediated motility in CFT073 is regulated by lpdA , which contributes to the understanding of how uropathogens differentially regulate motility mechanisms in the iron-restricted host. Importance Urinary tract infections (UTIs) are ubiquitous and responsible for over five billion dollars in associated health care costs annually. Both iron acquisition and motility are highly studied virulence factors associated with uropathogenic E. coli (UPEC), the main causative agent of uncomplicated UTI. This work is innovative by providing mechanistic insight into the synergistic relationship between these two critical virulence properties. Here, we demonstrate that iron limitation has pleiotropic effects with consequences that extend beyond metabolism, and impact other virulence mechanisms. Indeed, targeting iron acquisition as a therapy may lead to an undesirable enhancement of UPEC pathogenesis through increased motility. It is vital to understand the full breadth of UPEC pathogenesis to adequately respond to this common infection, especially with the increase of antibiotic resistant pathogens.
Collapse
|
15
|
Whelan S, Lucey B, Finn K. Uropathogenic Escherichia coli (UPEC)-Associated Urinary Tract Infections: The Molecular Basis for Challenges to Effective Treatment. Microorganisms 2023; 11:2169. [PMID: 37764013 PMCID: PMC10537683 DOI: 10.3390/microorganisms11092169] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Urinary tract infections (UTIs) are among the most common bacterial infections, especially among women and older adults, leading to a significant global healthcare cost burden. Uropathogenic Escherichia coli (UPEC) are the most common cause and accounts for the majority of community-acquired UTIs. Infection by UPEC can cause discomfort, polyuria, and fever. More serious clinical consequences can result in urosepsis, kidney damage, and death. UPEC is a highly adaptive pathogen which presents significant treatment challenges rooted in a complex interplay of molecular factors that allow UPEC to evade host defences, persist within the urinary tract, and resist antibiotic therapy. This review discusses these factors, which include the key genes responsible for adhesion, toxin production, and iron acquisition. Additionally, it addresses antibiotic resistance mechanisms, including chromosomal gene mutations, antibiotic deactivating enzymes, drug efflux, and the role of mobile genetic elements in their dissemination. Furthermore, we provide a forward-looking analysis of emerging alternative therapies, such as phage therapy, nano-formulations, and interventions based on nanomaterials, as well as vaccines and strategies for immunomodulation. This review underscores the continued need for research into the molecular basis of pathogenesis and antimicrobial resistance in the treatment of UPEC, as well as the need for clinically guided treatment of UTIs, particularly in light of the rapid spread of multidrug resistance.
Collapse
Affiliation(s)
- Shane Whelan
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland;
| | - Brigid Lucey
- Department of Biological Sciences, Munster Technological University, Bishopstown, T12 P928 Cork, Ireland;
| | - Karen Finn
- Department of Analytical, Biopharmaceutical and Medical Sciences, Atlantic Technological University Galway City, Dublin Road, H91 T8NW Galway, Ireland
| |
Collapse
|
16
|
Azam MW, Zarrilli R, Khan AU. Updates on the Virulence Factors Produced by Multidrug-Resistant Enterobacterales and Strategies to Control Their Infections. Microorganisms 2023; 11:1901. [PMID: 37630461 PMCID: PMC10456890 DOI: 10.3390/microorganisms11081901] [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: 03/15/2023] [Revised: 07/06/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
The Enterobacterales order is a massive group of Gram-negative bacteria comprised of pathogenic and nonpathogenic members, including beneficial commensal gut microbiota. The pathogenic members produce several pathogenic or virulence factors that enhance their pathogenic properties and increase the severity of the infection. The members of Enterobacterales can also develop resistance against the common antimicrobial agents, a phenomenon called antimicrobial resistance (AMR). Many pathogenic Enterobacterales members are known to possess antimicrobial resistance. This review discusses the virulence factors, pathogenicity, and infections caused by multidrug-resistant Enterobacterales, especially E. coli and some other bacterial species sharing similarities with the Enterobacterales members. We also discuss both conventional and modern approaches used to combat the infections caused by them. Understanding the virulence factors produced by the pathogenic bacteria will help develop novel strategies and methods to treat infections caused by them.
Collapse
Affiliation(s)
- Mohd W. Azam
- Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| | - Raffaele Zarrilli
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy
| | - Asad U. Khan
- Medical Microbiology and Molecular Biology Laboratory, Interdisciplinary Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, India
| |
Collapse
|
17
|
Kumari S, Howlett TS, Ehrman RN, Koirala S, Trashi O, Trashi I, Wijesundara YH, Gassensmith JJ. In vivo biocompatibility of ZIF-8 for slow release via intranasal administration. Chem Sci 2023; 14:5774-5782. [PMID: 37265713 PMCID: PMC10231336 DOI: 10.1039/d3sc00500c] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/28/2023] [Indexed: 06/03/2023] Open
Abstract
Zeolitic imidazolate framework-8 (ZIF-8) is becoming popular in research for its potential in antigen protection and for providing a thermally stable, slow-release platform. While papers applying this material for immunological applications are aplenty in the literature, studies that explore the biosafety of ZIF-8 in mammals-especially when administered intranasally-are not well represented. We checked the body clearance of uncoated and ZIF-8-coated liposomes and observed that the release slowed as ZIF-8 is easily degraded by mucosal fluid in the nasal cavity. We delivered varying doses of ZIF-8, checked its short- and long-term effects on diagnostic proteins found in blood serum, and found no noticeable differences from the saline control group. We also studied their lung diffusing capacity and tissue morphology; neither showed significant changes in morphology or function.
Collapse
Affiliation(s)
- Sneha Kumari
- Department of Chemistry and Biochemistry, The University of Texas at Dallas 800 West Campbell Rd. Richardson TX 75080 USA
| | - Thomas S Howlett
- Department of Chemistry and Biochemistry, The University of Texas at Dallas 800 West Campbell Rd. Richardson TX 75080 USA
| | - Ryanne N Ehrman
- Department of Chemistry and Biochemistry, The University of Texas at Dallas 800 West Campbell Rd. Richardson TX 75080 USA
| | - Shailendra Koirala
- Department of Chemistry and Biochemistry, The University of Texas at Dallas 800 West Campbell Rd. Richardson TX 75080 USA
| | - Orikeda Trashi
- Department of Chemistry and Biochemistry, The University of Texas at Dallas 800 West Campbell Rd. Richardson TX 75080 USA
| | - Ikeda Trashi
- Department of Chemistry and Biochemistry, The University of Texas at Dallas 800 West Campbell Rd. Richardson TX 75080 USA
| | - Yalini H Wijesundara
- Department of Chemistry and Biochemistry, The University of Texas at Dallas 800 West Campbell Rd. Richardson TX 75080 USA
| | - Jeremiah J Gassensmith
- Department of Chemistry and Biochemistry, The University of Texas at Dallas 800 West Campbell Rd. Richardson TX 75080 USA
- Department of Biomedical Engineering, The University of Texas at Dallas 800 West Campbell Rd. Richardson TX 75080 USA
| |
Collapse
|
18
|
Xing Y, Clark JR, Chang JD, Chirman DM, Green S, Zulk JJ, Jelinski J, Patras KA, Maresso AW. Broad protective vaccination against systemic Escherichia coli with autotransporter antigens. PLoS Pathog 2023; 19:e1011082. [PMID: 36800400 PMCID: PMC9937491 DOI: 10.1371/journal.ppat.1011082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 12/26/2022] [Indexed: 02/18/2023] Open
Abstract
Extraintestinal pathogenic Escherichia coli (ExPEC) is the leading cause of adult life-threatening sepsis and urinary tract infections (UTI). The emergence and spread of multidrug-resistant (MDR) ExPEC strains result in a considerable amount of treatment failure and hospitalization costs, and contribute to the spread of drug resistance amongst the human microbiome. Thus, an effective vaccine against ExPEC would reduce morbidity and mortality and possibly decrease carriage in healthy or diseased populations. A comparative genomic analysis demonstrated a gene encoding an invasin-like protein, termed sinH, annotated as an autotransporter protein, shows high prevalence in various invasive ExPEC phylogroups, especially those associated with systemic bacteremia and UTI. Here, we evaluated the protective efficacy and immunogenicity of a recombinant SinH-based vaccine consisting of either domain-3 or domains-1,2, and 3 of the putative extracellular region of surface-localized SinH. Immunization of a murine host with SinH-based antigens elicited significant protection against various strains of the pandemic ExPEC sequence type 131 (ST131) as well as multiple sequence types in two distinct models of infection (colonization and bacteremia). SinH immunization also provided significant protection against ExPEC colonization in the bladder in an acute UTI model. Immunized cohorts produced significantly higher levels of vaccine-specific serum IgG and urinary IgG and IgA, findings consistent with mucosal protection. Collectively, these results demonstrate that autotransporter antigens such as SinH may constitute promising ExPEC phylogroup-specific and sequence-type effective vaccine targets that reduce E. coli colonization and virulence.
Collapse
Affiliation(s)
- Yikun Xing
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - Justin R. Clark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - James D. Chang
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - Dylan M. Chirman
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - Sabrina Green
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - Jacob J. Zulk
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - Joseph Jelinski
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| | - Kathryn A. Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, Texas, United States of America
| | - Anthony W. Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
- TAILOR Labs, Vaccine Development Group, Baylor College of Medicine, Houston, Texas, United States of America
| |
Collapse
|
19
|
Pokharel P, Dhakal S, Dozois CM. The Diversity of Escherichia coli Pathotypes and Vaccination Strategies against This Versatile Bacterial Pathogen. Microorganisms 2023; 11:344. [PMID: 36838308 PMCID: PMC9965155 DOI: 10.3390/microorganisms11020344] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Escherichia coli (E. coli) is a gram-negative bacillus and resident of the normal intestinal microbiota. However, some E. coli strains can cause diseases in humans, other mammals and birds ranging from intestinal infections, for example, diarrhea and dysentery, to extraintestinal infections, such as urinary tract infections, respiratory tract infections, meningitis, and sepsis. In terms of morbidity and mortality, pathogenic E. coli has a great impact on public health, with an economic cost of several billion dollars annually worldwide. Antibiotics are not usually used as first-line treatment for diarrheal illness caused by E. coli and in the case of bloody diarrhea, antibiotics are avoided due to the increased risk of hemolytic uremic syndrome. On the other hand, extraintestinal infections are treated with various antibiotics depending on the site of infection and susceptibility testing. Several alarming papers concerning the rising antibiotic resistance rates in E. coli strains have been published. The silent pandemic of multidrug-resistant bacteria including pathogenic E. coli that have become more difficult to treat favored prophylactic approaches such as E. coli vaccines. This review provides an overview of the pathogenesis of different pathotypes of E. coli, the virulence factors involved and updates on the major aspects of vaccine development against different E. coli pathotypes.
Collapse
Affiliation(s)
- Pravil Pokharel
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal Saint-Hyacinthe, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Sabin Dhakal
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal Saint-Hyacinthe, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Charles M. Dozois
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal Saint-Hyacinthe, Saint-Hyacinthe, QC J2S 2M2, Canada
- Pasteur Network, Laval, QC H7V 1B7, Canada
| |
Collapse
|
20
|
Ligon MM, Joshi CS, Fashemi BE, Salazar AM, Mysorekar IU. Effects of aging on urinary tract epithelial homeostasis and immunity. Dev Biol 2023; 493:29-39. [PMID: 36368522 PMCID: PMC11463731 DOI: 10.1016/j.ydbio.2022.11.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
A global increase in older individuals creates an increasing demand to understand numerous healthcare challenges related to aging. This population is subject to changes in tissue physiology and the immune response network. Older individuals are particularly susceptible to infectious diseases, with one of the most common being urinary tract infections (UTIs). Postmenopausal and older women have the highest risk of recurrent UTIs (rUTIs); however, why rUTIs become more frequent after menopause and during old age is incompletely understood. This increased susceptibility and severity among older individuals may involve functional changes to the immune system with age. Aging also has substantial effects on the epithelium and the immune system that led to impaired protection against pathogens, yet heightened and prolonged inflammation. How the immune system and its responses to infection changes within the bladder mucosa during aging has largely remained poorly understood. In this review, we highlight our understanding of bladder innate and adaptive immunity and the impact of aging and hormones and hormone therapy on bladder epithelial homeostasis and immunity. In particular, we elaborate on how the cellular and molecular immune landscape within the bladder can be altered during aging as aged mice develop bladder tertiary lymphoid tissues (bTLT), which are absent in young mice leading to profound age-associated change to the immune landscape in bladders that might drive the significant increase in UTI susceptibility. Knowledge of host factors that prevent or promote infection can lead to targeted treatment and prevention regimens. This review also identifies unique host factors to consider in the older, female host for improving rUTI treatment and prevention by dissecting the age-associated alteration of the bladder mucosal immune system.
Collapse
Affiliation(s)
- Marianne M Ligon
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Chetanchandra S Joshi
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Bisiayo E Fashemi
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Arnold M Salazar
- Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Indira U Mysorekar
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, MO, 63110, USA; Department of Medicine, Section of Infectious Diseases, Baylor College of Medicine, Houston, TX, 77030, USA; Department of Molecular Microbiology and Virology, Baylor College of Medicine, Houston, TX, 77030, USA; Huffington Center on Aging, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
21
|
Ribeiro-Filho L, Suartz CV, Braz NDSF, Siqueira M, Hirasaki F, Mitre AI, Gomes CM, Nahas WC. Long-term efficacy of complete trigonal electrofulguration for women with recurrent urinary tract infections. Neurourol Urodyn 2023; 42:188-193. [PMID: 36321758 DOI: 10.1002/nau.25066] [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: 08/09/2022] [Revised: 09/25/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION AND OBJECTIVE Recurrent urinary tract infections (R-UTIs) have a negative impact on quality of life and contribute to antimicrobial resistance. Long-term antibiotic therapy is the main treatment alternative but, in some cases, this approach may not be accepted by the patient, is ineffective or poorly tolerated. In selected women, electrofulguration (EF) of trigonal lesions has been shown to reduce urinary tract infection (UTI) episodes. METHODS Between August 2006 and December 2017, 73 women with R-UTI had their data collected prospectively and analyzed. We evaluated the rate of UTIs during the initial 2 years of follow-up after fulguration based on symptoms and a positive urine culture. All patients failed with multiple antibiotic courses and were offered endoscopic electrofulguration of the entire trigonal and bladder neck mucosa with a rollerball probe. We present our long-term results using a strategy of complete fulguration of the trigone in women with R-UTIs. RESULTS The median age was 64 years (range: 17-76 years) and the median follow-up time after EF was 4.2 years (range: 2.5-14 years). Overall, 70 patients (96%) remained free of UTI episodes during the first year of follow-up, at 2 years of follow-up, 57, 53% remained infection-free. Currently, UTIs are typically sparse, mild, and caused by multisensitive bacteria. CONCLUSIONS Complete trigonal and bladder neck mucosal fulguration promoted a significant reduction of UTI episodes during the first 2 years of follow-up. Prospective controlled studies are needed to determine the role of EF in women with R-UTI.
Collapse
Affiliation(s)
| | - Caio V Suartz
- Division of Urology, University of São Paulo School of Medicine, Sao Paulo, Brazil
| | - Natalia D S F Braz
- Division of Urology, University of São Paulo School of Medicine, Sao Paulo, Brazil
| | - Matheus Siqueira
- Division of Urology, University of São Paulo School of Medicine, Sao Paulo, Brazil
| | - Felipe Hirasaki
- Division of Urology, University of São Paulo School of Medicine, Sao Paulo, Brazil
| | - Anuar I Mitre
- Division of Urology, University of São Paulo School of Medicine, Sao Paulo, Brazil
| | - Cristiano M Gomes
- Division of Urology, University of São Paulo School of Medicine, Sao Paulo, Brazil
| | - William C Nahas
- Division of Urology, University of São Paulo School of Medicine, Sao Paulo, Brazil
| |
Collapse
|
22
|
Kelly SH, Votaw NL, Cossette BJ, Wu Y, Shetty S, Shores LS, Issah LA, Collier JH. A sublingual nanofiber vaccine to prevent urinary tract infections. SCIENCE ADVANCES 2022; 8:eabq4120. [PMID: 36417519 PMCID: PMC9683704 DOI: 10.1126/sciadv.abq4120] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 09/21/2022] [Indexed: 06/16/2023]
Abstract
Urinary tract infections (UTIs) are a major public health problem affecting millions of individuals each year. Recurrent UTIs are managed by long-term antibiotic use, making the alarming rise of antibiotic resistance a substantial threat to future UTI treatment. Extended antibiotic regimens may also have adverse effects on the microbiome. Here, we report the use of a supramolecular vaccine to provide long-term protection against uropathogenic Escherichia coli, which cause 80% of uncomplicated UTIs. We designed mucus-penetrating peptide-polymer nanofibers to enable sublingual (under the tongue) vaccine delivery and elicit antibody responses systemically and in the urogenital tract. In a mouse model of UTI, we demonstrate equivalent efficacy to high-dose oral antibiotics but with significantly less perturbation of the gut microbiome. We also formulate our vaccine as a rapid-dissolving sublingual tablet that raises response in mice and rabbits. Our approach represents a promising alternative to antibiotics for the treatment and prevention of UTIs.
Collapse
Affiliation(s)
| | - Nicole L. Votaw
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Yaoying Wu
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Shamitha Shetty
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Luqman A. Issah
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | |
Collapse
|
23
|
Geurtsen J, de Been M, Weerdenburg E, Zomer A, McNally A, Poolman J. Genomics and pathotypes of the many faces of Escherichia coli. FEMS Microbiol Rev 2022; 46:fuac031. [PMID: 35749579 PMCID: PMC9629502 DOI: 10.1093/femsre/fuac031] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 06/22/2022] [Indexed: 01/09/2023] Open
Abstract
Escherichia coli is the most researched microbial organism in the world. Its varied impact on human health, consisting of commensalism, gastrointestinal disease, or extraintestinal pathologies, has generated a separation of the species into at least eleven pathotypes (also known as pathovars). These are broadly split into two groups, intestinal pathogenic E. coli (InPEC) and extraintestinal pathogenic E. coli (ExPEC). However, components of E. coli's infinite open accessory genome are horizontally transferred with substantial frequency, creating pathogenic hybrid strains that defy a clear pathotype designation. Here, we take a birds-eye view of the E. coli species, characterizing it from historical, clinical, and genetic perspectives. We examine the wide spectrum of human disease caused by E. coli, the genome content of the bacterium, and its propensity to acquire, exchange, and maintain antibiotic resistance genes and virulence traits. Our portrayal of the species also discusses elements that have shaped its overall population structure and summarizes the current state of vaccine development targeted at the most frequent E. coli pathovars. In our conclusions, we advocate streamlining efforts for clinical reporting of ExPEC, and emphasize the pathogenic potential that exists throughout the entire species.
Collapse
Affiliation(s)
- Jeroen Geurtsen
- Janssen Vaccines and Prevention B.V., 2333 Leiden, the Netherlands
| | - Mark de Been
- Janssen Vaccines and Prevention B.V., 2333 Leiden, the Netherlands
| | | | - Aldert Zomer
- Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, 3584 Utrecht, the Netherlands
| | - Alan McNally
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, B15 2TT Birmingham, United Kingdom
| | - Jan Poolman
- Janssen Vaccines and Prevention B.V., 2333 Leiden, the Netherlands
| |
Collapse
|
24
|
Tobuse AJ, Ang CW, Yeong KY. Modern vaccine development via reverse vaccinology to combat antimicrobial resistance. Life Sci 2022; 302:120660. [PMID: 35642852 DOI: 10.1016/j.lfs.2022.120660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/02/2022] [Accepted: 05/19/2022] [Indexed: 10/18/2022]
Abstract
With the continuous evolution of bacteria, the global antimicrobial resistance health threat is causing millions of deaths yearly. While depending on antibiotics as a primary treatment has its merits, there are no effective alternatives thus far in the pharmaceutical market against some drug-resistant bacteria. In recent years, vaccinology has become a key topic in scientific research. Combining with the growth of technology, vaccine research is seeing a new light where the process is made faster and more efficient. Although less discussed, bacterial vaccine is a feasible strategy to combat antimicrobial resistance. Some vaccines have shown promising results with good efficacy against numerous multidrug-resistant strains of bacteria. In this review, we aim to discuss the findings from studies utilizing reverse vaccinology for vaccine development against some multidrug-resistant bacteria, as well as provide a summary of multi-year bacterial vaccine studies in clinical trials. The advantages of reverse vaccinology in the generation of new bacterial vaccines are also highlighted. Meanwhile, the limitations and future prospects of bacterial vaccine concludes this review.
Collapse
Affiliation(s)
- Asuka Joy Tobuse
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia
| | - Chee Wei Ang
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia
| | - Keng Yoon Yeong
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway 47500, Selangor, Malaysia.
| |
Collapse
|
25
|
Butler D, Ambite I, Wan MLY, Tran TH, Wullt B, Svanborg C. Immunomodulation therapy offers new molecular strategies to treat UTI. Nat Rev Urol 2022; 19:419-437. [PMID: 35732832 PMCID: PMC9214477 DOI: 10.1038/s41585-022-00602-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2022] [Indexed: 12/13/2022]
Abstract
Innovative solutions are needed for the treatment of bacterial infections, and a range of antibacterial molecules have been explored as alternatives to antibiotics. A different approach is to investigate the immune system of the host for new ways of making the antibacterial defence more efficient. However, the immune system has a dual role as protector and cause of disease: in addition to being protective, increasing evidence shows that innate immune responses can become excessive and cause acute symptoms and tissue pathology during infection. This role of innate immunity in disease suggests that the immune system should be targeted therapeutically, to inhibit over-reactivity. The ultimate goal is to develop therapies that selectively attenuate destructive immune response cascades, while augmenting the protective antimicrobial defence but such treatment options have remained underexplored, owing to the molecular proximity of the protective and destructive effects of the immune response. The concept of innate immunomodulation therapy has been developed successfully in urinary tract infections, based on detailed studies of innate immune activation and disease pathogenesis. Effective, disease-specific, immunomodulatory strategies have been developed by targeting specific immune response regulators including key transcription factors. In acute pyelonephritis, targeting interferon regulatory factor 7 using small interfering RNA or treatment with antimicrobial peptide cathelicidin was protective and, in acute cystitis, targeting overactive effector molecules such as IL-1β, MMP7, COX2, cAMP and the pain-sensing receptor NK1R has been successful in vivo. Furthermore, other UTI treatment strategies, such as inhibiting bacterial adhesion and vaccination, have also shown promise. Hyperactivation of innate immunity is a disease determinant in urinary tract infections (UTIs). Modulation of innate immunity has promise as a therapy for UTIs. In this Review, the authors discuss potential mechanisms and immunomodulatory therapeutic strategies in UTIs. Excessive innate immune responses to infection cause symptoms and pathology in acute pyelonephritis and acute cystitis. Innate immunomodulation therapy is, therefore, a realistic option for treating these conditions. Targeting excessive innate immune responses at the level of transcription has been successful in animal models. Innate immunomodulation therapy reduces excessive inflammation and tissue pathology and accelerates bacterial clearance from infected kidneys and bladders in mice. Innate immunomodulation therapy also accelerates the clearance of antibiotic-resistant bacterial strains.
Collapse
Affiliation(s)
- Daniel Butler
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ines Ambite
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Murphy Lam Yim Wan
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Thi Hien Tran
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Björn Wullt
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden
| | - Catharina Svanborg
- Department of Microbiology, Immunology and Glycobiology, Institute of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
26
|
Abstract
More than half of women will experience a urinary tract infection (UTI), with uropathogenic Escherichia coli (UPEC) causing ~80% of uncomplicated cases. Iron acquisition systems are essential for uropathogenesis, and UPEC strains encode highly diverse iron acquisition systems, underlining their importance. However, a recent UPEC clinical isolate, HM7, lacks this diversity and instead encodes the synthesis pathway for a sole siderophore, enterobactin. To determine if HM7 possesses unidentified iron acquisition systems, we performed RNA sequencing under iron-limiting conditions and demonstrated that the ferric citrate uptake system (fecABCDE and fecIR) was highly upregulated. Importantly, there are high levels of citrate within urine, some of which is bound to iron, and the fec system is enriched in UPEC isolates compared to fecal strains. Therefore, we hypothesized that HM7 and other similar strains use the fec system to acquire iron in the host. Deletion of both enterobactin biosynthesis and ferric citrate uptake (ΔfecA/ΔentB) abrogates use of ferric citrate as an iron source, and fecA provides an advantage in human urine in the absence of enterobactin. However, in a UTI mouse model, fecA is a fitness factor independent of enterobactin production, likely due to the action of host lipocalin-2 chelating ferrienterobactin. These findings indicate that ferric citrate uptake is used as an iron source when siderophore efficacy is limited, such as in the host during UTI. Defining these novel compensatory mechanisms and understanding the nutritional hierarchy of preferred iron sources within the urinary tract are important in the search for new approaches to combat UTI.
Collapse
|
27
|
Vaccination of mice with hybrid protein containing Exotoxin S and PcrV with adjuvants alum and MPL protects Pseudomonas aeruginosa infections. Sci Rep 2022; 12:1325. [PMID: 35079054 PMCID: PMC8789797 DOI: 10.1038/s41598-022-05157-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 01/07/2022] [Indexed: 11/28/2022] Open
Abstract
Pseudomonas aeruginosa as a common pathogen causing urinary tract infections (UTIs) has been resistant to different antibiotics and developing an effective vaccine can be an alternative strategy. In the present study, the immunogenicity and protection efficacy of formulations composed of a hybrid protein composed of P. aeruginosa V-antigen (PcrV) and exoenzyme S (ExoS) with alum and MPL were evaluated. The hybrid protein could increase the specific systemic and mucosal immune responses, as well as cellular responses as compared with control groups. Combining of alum or MPL adjuvant with the hybrid protein significantly improved the levels of IgG1, serum IgA, mucosal IgG, and IL-17 as compared to the ExoS.PcrV alone. After bladder challenge with a P. aeruginosa strain, the bacterial loads of bladder and kidneys were significantly decreased in mice received ExoS.PcrV admixed with alum and ExoS.PcrV admixed with MPL than controls. The present study indicated that immunization of mice with a hybrid protein composed of ExoS and PcrV could induce multifactorial immune responses and opsonize the bacteria and decrease the viable bacterial cells. Because P. aeruginosa have caused therapeutic challenges worldwide, our study proposed ExoS.PcrV + alum and ExoS.PcrV + MPL as promising candidates for the prevention of infections caused by P. aeruginosa.
Collapse
|
28
|
Chang Z, An L, He Z, Zhang Y, Li S, Lei M, Xu P, Lai Y, Jiang Z, Huang Y, Duan X, Wu W. Allicin supressed Escherichia coli-induced urinary tract infections by a Novel MALT1/NF-κB pathway. Food Funct 2022; 13:3495-3511. [PMID: 35246671 DOI: 10.1039/d1fo03853b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Escherichia coli (E. coli) strains cause the majority of urinary tract infections (UTIs) and are resistant to various antibiotics. Therefore, it is imperative to explore novel host-target therapies. As a...
Collapse
Affiliation(s)
- Zhenglin Chang
- Department of Urology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
- Department of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, 510230, China
| | - Lingyue An
- Department of Urology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
- Department of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, 510230, China
| | - Zhican He
- Department of Urology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
- Department of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, 510230, China
| | - Yuyan Zhang
- Guangzhou Institute of Dermatology, Guangzhou, 510095, China
| | - Shujue Li
- Department of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, 510230, China
| | - Min Lei
- Department of Urology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
- Department of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, 510230, China
| | - Peng Xu
- Department of Urology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
- Department of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, 510230, China
| | - Yongchang Lai
- Department of Urology, Shenzhen Shockwave Lithotripsy Research Institute, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, Guangdong 518033, China
| | - Zheng Jiang
- Department of Urology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
- Department of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, 510230, China
| | - Yapeng Huang
- Department of Urology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
- Department of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, 510230, China
| | - Xiaolu Duan
- Department of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, 510230, China
| | - Wenqi Wu
- Department of Urology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, 510260, China.
- Department of Urology, the First Affiliated Hospital of Guangzhou Medical University, Guangdong Key Laboratory of Urology, Guangzhou, 510230, China
| |
Collapse
|
29
|
Wenzel M, Hoeh B, Goeldner K, Preisser F, Würnschimmel C, Becker A, Mandel P, Karakiewicz PI, Chun FKH, Kluth LA. Catheterization Does Not Improve Course of Disease in Female Patients with Acute Cystitis or Pyelonephritis: Retrospective Analysis of >300 In-Hospital Treated Patients. Urol Int 2021; 105:1104-1112. [PMID: 34515228 DOI: 10.1159/000518066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 06/22/2021] [Indexed: 11/19/2022]
Abstract
PURPOSE Females with in-hospital treatment for acute cystitis (AC) or pyelonephritis may benefit from catheterization at admission. METHODS All female patients with AC or pyelonephritis requiring in-hospital treatment at University Hospital Frankfurt (2004-2019) were retrospectively analyzed. Logistic regression models were used to predict the catheter value. RESULTS Of 310 female patients, 40% harbored AC versus 60% pyelonephritis, of whom 62% and 74% received a catheter at admission: C-reactive protein (CRP) and white blood count (WBC) were significantly elevated in AC and pyelonephritis catheter versus no catheter patients (both p < 0.05). Time to CRP and WBC nadir did not differ between the AC catheter versus no catheter group (both p > 0.05). Conversely, time to CRP nadir was prolonged in pyelonephritis catheter patients. AC and pyelonephritis catheter patients exhibited a prolonged antibiotic treatment and length of stay (LOS, both p < 0.05). In multivariable analyses, CRP >5 ng/mL was a predictor for receiving a catheter in all patients. In AC, a positive urine culture and fever predicted, respectively, prolonged LOS or antibiotic treatment (all p < 0.05). CONCLUSION Risk factors exist with regard to receiving a catheter and prolonged antibiotic treatment or LOS in females with AC or pyelonephritis. A catheter may not accelerate recovery or WBC nadir.
Collapse
Affiliation(s)
- Mike Wenzel
- Department of Urology, University Hospital Frankfurt, Frankfurt, Germany.,Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
| | - Benedikt Hoeh
- Department of Urology, University Hospital Frankfurt, Frankfurt, Germany.,Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
| | - Konstatin Goeldner
- Department of Urology, University Hospital Frankfurt, Frankfurt, Germany
| | - Felix Preisser
- Department of Urology, University Hospital Frankfurt, Frankfurt, Germany
| | - Christoph Würnschimmel
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas Becker
- Department of Urology, University Hospital Frankfurt, Frankfurt, Germany
| | - Philipp Mandel
- Department of Urology, University Hospital Frankfurt, Frankfurt, Germany
| | - Pierre I Karakiewicz
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
| | - Felix K-H Chun
- Department of Urology, University Hospital Frankfurt, Frankfurt, Germany
| | - Luis A Kluth
- Department of Urology, University Hospital Frankfurt, Frankfurt, Germany
| |
Collapse
|
30
|
Peck J, Shepherd JP. Recurrent Urinary Tract Infections: Diagnosis, Treatment, and Prevention. Obstet Gynecol Clin North Am 2021; 48:501-513. [PMID: 34416934 DOI: 10.1016/j.ogc.2021.05.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Urinary tract infections are common infections seen by obstetrician/gynecologists and primary care providers. Recurrent urinary tract infections are difficult to treat and decrease quality of life. This article addresses the diagnosis, treatment, and prevention of recurrent urinary tract infections, including pharmacologic and nonpharmacologic interventions. Most notably, increased interest in the urinary microbiome is leading to improved understanding of virulent and commensal bacteria in the genitourinary tract. Understanding the causative factors of recurrent urinary tract infections will lead to improved detection, treatment, and prevention.
Collapse
Affiliation(s)
- Julie Peck
- St Francis Hospital Trinity Health, 114 Woodland Street, OB Administration 43, Hartford, CT 06105, USA.
| | - Jonathan P Shepherd
- St Francis Hospital Trinity Health, 114 Woodland Street, OB Administration 43, Hartford, CT 06105, USA
| |
Collapse
|
31
|
Local induction of bladder Th1 responses to combat urinary tract infections. Proc Natl Acad Sci U S A 2021; 118:2026461118. [PMID: 33653961 DOI: 10.1073/pnas.2026461118] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Given the high frequency of urinary tract infections (UTIs) and their recurrence, there is keen interest in developing effective UTI vaccines. Currently, most vaccine studies, including those in humans, involve parenteral vaccination aimed at evoking and sustaining elevated levels of systemic antibody directed at the uropathogens. In view of recent reports of aberrant Th2-biased bladder immune responses to infection, we hypothesized that immunizing mice intravesically with antigens from uropathogenic Escherichia coli (UPEC) combined with a Th1-skewing adjuvant could correct this defect and promote protection against UTIs. Here we report that compared with mice immunized subcutaneously with this vaccine combination, intravesically immunized mice were markedly more protected from UTIs because of their distinctive ability to recruit Th1 cells into the bladder. This mode of vaccination was effective even in mice that experienced multiple UTIs and displayed pronounced aberrant bladder immune responses. Thus, intravesical vaccination with one or more UPEC antigens to induce bladder Th1 responses represents a superior strategy to combat UTIs, especially in UTI-prone subjects.
Collapse
|
32
|
Comparative Pathogenomics of Escherichia coli: Polyvalent Vaccine Target Identification through Virulome Analysis. Infect Immun 2021; 89:e0011521. [PMID: 33941580 PMCID: PMC8281228 DOI: 10.1128/iai.00115-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Comparative genomics of bacterial pathogens has been useful for revealing potential virulence factors. Escherichia coli is a significant cause of human morbidity and mortality worldwide but can also exist as a commensal in the human gastrointestinal tract. With many sequenced genomes, it has served as a model organism for comparative genomic studies to understand the link between genetic content and potential for virulence. To date, however, no comprehensive analysis of its complete “virulome” has been performed for the purpose of identifying universal or pathotype-specific targets for vaccine development. Here, we describe the construction of a pathotype database of 107 well-characterized completely sequenced pathogenic and nonpathogenic E. coli strains, which we annotated for major virulence factors (VFs). The data are cross referenced for patterns against pathotype, phylogroup, and sequence type, and the results were verified against all 1,348 complete E. coli chromosomes in the NCBI RefSeq database. Our results demonstrate that phylogroup drives many of the “pathotype-associated” VFs, and ExPEC-associated VFs are found predominantly within the B2/D/F/G phylogenetic clade, suggesting that these phylogroups are better adapted to infect human hosts. Finally, we used this information to propose polyvalent vaccine targets with specificity toward extraintestinal strains, targeting key invasive strategies, including immune evasion (group 2 capsule), iron acquisition (FyuA, IutA, and Sit), adherence (SinH, Afa, Pap, Sfa, and Iha), and toxins (Usp, Sat, Vat, Cdt, Cnf1, and HlyA). While many of these targets have been proposed before, this work is the first to examine their pathotype and phylogroup distribution and how they may be targeted together to prevent disease.
Collapse
|
33
|
Habibi M, Azimi S, Khoobbakht D, Roghanian P, Asadi Karam MR. Immunization with recombinant protein Ag43::UpaH with alum and 1,25(OH)2D3 adjuvants significantly protects Balb/C mice against urinary tract infection caused by uropathogenic Escherichia coli. Int Immunopharmacol 2021; 96:107638. [PMID: 33848909 DOI: 10.1016/j.intimp.2021.107638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/31/2021] [Accepted: 03/31/2021] [Indexed: 11/29/2022]
Abstract
The majority of urinary tract infections (UTIs) are caused by uropathogenic Escherichia coli (UPEC). Designing a vaccine will certainly reduce the occurrence of infection and antibiotic resistance of the isolates. Antigen 43 (Ag43) and autotransporter H (UpaH) have been associated with the virulence of UPEC. In the present study, the efficacy of different formulations of a hybrid protein composed of Ag43 and UpaH with and without alum and 1,25(OH)2D3 (Vitamin D3) adjuvants were evaluated in mice model. A significant increase in IgG and cellular responses was developed against Ag43::UpaH as compared to the control mice. The addition of alum or a mixture of alum and Vitamin D3 to the protein significantly enhanced the serum IgG responses and tended to remain in a steady state until 6 months. In addition, the mentioned formulations produced significant amounts of IgG1, IL-4, and IL-17 as compared to the fusion protein alone. In addition to the mentioned formulations, the combination of protein with Vitamin D3 also resulted in significantly higher serum IgA and IFN-γ levels as compared to the fusion protein alone. Mice immunized with fusion plus alum and formulation protein admixed with both alum and Vitamin D3 significantly reduced the bacterial load in the bladders and kidneys of mice as compared to the control. In this study, for the first time, the ability of a novel hybrid protein in combination with adjuvants alum and Vitamin D3 was evaluated against UPEC. Our results indicated that fusion Ag43::UpaH admixed with alum and Vitamin D3 could be a promising candidate against UTIs.
Collapse
Affiliation(s)
- Mehri Habibi
- Department of Molecular Biology, Pasteur Institute of Iran, Pasteur Ave, Tehran 13164, Iran
| | - Saba Azimi
- Department of Molecular Biology, Pasteur Institute of Iran, Pasteur Ave, Tehran 13164, Iran
| | - Dorna Khoobbakht
- Department of Molecular Biology, Pasteur Institute of Iran, Pasteur Ave, Tehran 13164, Iran
| | - Pooneh Roghanian
- Department of Molecular Biology, Pasteur Institute of Iran, Pasteur Ave, Tehran 13164, Iran
| | | |
Collapse
|
34
|
The prevalence of the iutA and ibeA genes in Escherichia coli isolates from severe and non-severe patients with bacteremic acute biliary tract infection is significantly different. Gut Pathog 2021; 13:32. [PMID: 34006312 PMCID: PMC8132388 DOI: 10.1186/s13099-021-00429-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 05/12/2021] [Indexed: 12/17/2022] Open
Abstract
Background Although Escherichia coli is the most frequently isolated microorganism in acute biliary tract infections with bacteremia, data regarding its virulence are limited. Results Information on cases of bacteremia in acute biliary tract infection in a retrospective study was collected from 2013 to 2015 at a tertiary care hospital in Japan. Factors related to the severity of infection were investigated, including patient background, phylogenetic typing, and virulence factors of E. coli, such as adhesion, invasion, toxins, and iron acquisition. In total, 72 E. coli strains were identified in 71 cases, most of which primarily belonged to the B2 phylogroup (68.1%). The presence of the iutA gene (77.3% in the non-severe group, 46.4% in the severe group, P = 0.011) and the ibeA gene (9.1% in the non-severe group, and 35.7% in the severe group, P = 0.012) was significantly associated with the severity of infection. Among the patient characteristics, diabetes mellitus with organ involvement and alkaline phosphatase were different in the severe and non-severe groups. Conclusions We showed that bacteremic E. coli strains from acute biliary tract infections belonged to the virulent (B2) phylogroup. The prevalence of the iutA and ibeA genes between the two groups of bacteremia severity was significantly different.
Collapse
|
35
|
Karan S, Garg LC, Choudhury D, Dixit A. Recombinant FimH, a fimbrial tip adhesin of Vibrio parahaemolyticus, elicits mixed T helper cell response and confers protection against Vibrio parahaemolyticus challenge in murine model. Mol Immunol 2021; 135:373-387. [PMID: 34020083 DOI: 10.1016/j.molimm.2021.05.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/21/2021] [Accepted: 05/09/2021] [Indexed: 11/18/2022]
Abstract
Vibrio parahaemolyticus causes vibriosis in wide range of marine organisms, and is responsible for food borne illnesses in humans through consumption of contaminated uncooked/partially cooked seafood. Continued and widespread antibiotics usage to increase the productivity has led to antibiotics resistance development. This has necessitated the need to develop alternative methods to control its infection. Use of safe and effective vaccines against the virulence factors not only protects from infection, it also minimizes antibiotic usage. The colonization of V. parahaemolyticus in the host and disease development requires several adhesins present on the cell surface, and thereby make them attractive vaccine candidates. V. parahaemolyticus produces extracellular type 1 fimbriae that have been shown to play a role in adhesion, biofilm formation and virulence. FimH is one of the minor components of the type 1 fimbriae occurring on its very tip. Being present on the cell surface, it is highly immunogenic, and can be targeted as a potential vaccine candidate. The present study describes the immunogenic and vaccine potential of recombinant V. parahaemolyticus FimH (rVpFimH) expressed in E. coli. Immunization of BALB/c mice with the rVpFimH elicited a strong mixed immune response, T-cell memory (evidenced by antibody isotyping, cytokine profiling and T-cell proliferation assay), and agglutination positive antibodies. FACS analysis and immunogold labeling showed that the polyclonal anti-rVpFimH antibodies were able to recognize the FimH on V. parahaemolyticus cells. In vivo challenge of the rVpFimH-immunized mice with 2×LD50 dose of live bacteria showed one hundred percent survival. Thus, our findings clearly demonstrate the potential of FimH as an effective vaccine candidate against V. parahaemolyticus.
Collapse
Affiliation(s)
- Sweta Karan
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Lalit C Garg
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, 110067, India
| | - Devapriya Choudhury
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Aparna Dixit
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
36
|
Avian Pathogenic Escherichia coli (APEC): An Overview of Virulence and Pathogenesis Factors, Zoonotic Potential, and Control Strategies. Pathogens 2021; 10:pathogens10040467. [PMID: 33921518 PMCID: PMC8069529 DOI: 10.3390/pathogens10040467] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/05/2021] [Accepted: 04/09/2021] [Indexed: 02/07/2023] Open
Abstract
Avian pathogenic Escherichia coli (APEC) causes colibacillosis in avian species, and recent reports have suggested APEC as a potential foodborne zoonotic pathogen. Herein, we discuss the virulence and pathogenesis factors of APEC, review the zoonotic potential, provide the current status of antibiotic resistance and progress in vaccine development, and summarize the alternative control measures being investigated. In addition to the known virulence factors, several other factors including quorum sensing system, secretion systems, two-component systems, transcriptional regulators, and genes associated with metabolism also contribute to APEC pathogenesis. The clear understanding of these factors will help in developing new effective treatments. The APEC isolates (particularly belonging to ST95 and ST131 or O1, O2, and O18) have genetic similarities and commonalities in virulence genes with human uropathogenic E. coli (UPEC) and neonatal meningitis E. coli (NMEC) and abilities to cause urinary tract infections and meningitis in humans. Therefore, the zoonotic potential of APEC cannot be undervalued. APEC resistance to almost all classes of antibiotics, including carbapenems, has been already reported. There is a need for an effective APEC vaccine that can provide protection against diverse APEC serotypes. Alternative therapies, especially the virulence inhibitors, can provide a novel solution with less likelihood of developing resistance.
Collapse
|
37
|
Vaccines against antimicrobial resistance: a promising escape route for multidrug resistance. Pharm Pat Anal 2021; 10:83-98. [PMID: 33829866 DOI: 10.4155/ppa-2020-0022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Antibiotic resistance has become a global health problem requiring urgent intervention. The pace of development and frequency of transmission of antimicrobial resistance have tremendously surpassed the number of antibiotics developed in the past few decades. Emergence and transmission of multidrug-resistant genes, for example, mcr-1 and mcr-5.3, against the last resort of antibiotics has challenged the treatment options. Vaccination is a promising approach with no instance of antimicrobial resistance generation or transmission reported so far. The time required for developing a vaccine, extensive pre- and post-licensure studies and the financial constraints for the R&D has hampered vaccine development over the past few decades. Vaccine can prove to be an effective future strategy for combating antimicrobial resistance.
Collapse
|
38
|
Sargun A, Johnstone TC, Zhi H, Raffatellu M, Nolan EM. Enterobactin- and salmochelin-β-lactam conjugates induce cell morphologies consistent with inhibition of penicillin-binding proteins in uropathogenic Escherichia coli CFT073. Chem Sci 2021; 12:4041-4056. [PMID: 34163675 PMCID: PMC8179508 DOI: 10.1039/d0sc04337k] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 12/31/2020] [Indexed: 12/15/2022] Open
Abstract
The design and synthesis of narrow-spectrum antibiotics that target a specific bacterial strain, species, or group of species is a promising strategy for treating bacterial infections when the causative agent is known. In this work, we report the synthesis and evaluation of four new siderophore-β-lactam conjugates where the broad-spectrum β-lactam antibiotics cephalexin (Lex) and meropenem (Mem) are covalently attached to either enterobactin (Ent) or diglucosylated Ent (DGE) via a stable polyethylene glycol (PEG3) linker. These siderophore-β-lactam conjugates showed enhanced minimum inhibitory concentrations against Escherichia coli compared to the parent antibiotics. Uptake studies with uropathogenic E. coli CFT073 demonstrated that the DGE-β-lactams target the pathogen-associated catecholate siderophore receptor IroN. A comparative analysis of siderophore-β-lactams harboring ampicillin (Amp), Lex and Mem indicated that the DGE-Mem conjugate is advantageous because it targets IroN and exhibits low minimum inhibitory concentrations, fast time-kill kinetics, and enhanced stability to serine β-lactamases. Phase-contrast and fluorescence imaging of E. coli treated with the siderophore-β-lactam conjugates revealed cellular morphologies consistent with the inhibition of penicillin-binding proteins PBP3 (Ent/DGE-Amp/Lex) and PBP2 (Ent/DGE-Mem). Overall, this work illuminates the uptake and cell-killing activity of Ent- and DGE-β-lactam conjugates against E. coli and supports that native siderophore scaffolds provide the opportunity for narrowing the activity spectrum of antibiotics in clinical use and targeting pathogenicity.
Collapse
Affiliation(s)
- Artur Sargun
- Department of Chemistry, Massachusetts Institute of Technology Cambridge MA 02139 USA +1-617-452-2495
| | - Timothy C Johnstone
- Department of Chemistry, Massachusetts Institute of Technology Cambridge MA 02139 USA +1-617-452-2495
| | - Hui Zhi
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego La Jolla CA 92093 USA
| | - Manuela Raffatellu
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego La Jolla CA 92093 USA
- Center for Microbiome Innovation, University of California San Diego La Jolla CA 92093 USA
- Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines La Jolla CA 92093 USA
| | - Elizabeth M Nolan
- Department of Chemistry, Massachusetts Institute of Technology Cambridge MA 02139 USA +1-617-452-2495
| |
Collapse
|
39
|
Sargun A, Gerner RR, Raffatellu M, Nolan EM. Harnessing Iron Acquisition Machinery to Target Enterobacteriaceae. J Infect Dis 2020; 223:S307-S313. [PMID: 33330928 DOI: 10.1093/infdis/jiaa440] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Infections caused by Gram-negative bacteria can be challenging to treat due to the outer membrane permeability barrier and the increasing emergence of antibiotic resistance. During infection, Gram-negative pathogens must acquire iron, an essential nutrient, in the host. Many Gram-negative bacteria utilize sophisticated iron acquisition machineries based on siderophores, small molecules that bind iron with high affinity. In this review, we provide an overview of siderophore-mediated iron acquisition in Enterobacteriaceae and show how these systems provide a foundation for the conceptualization and development of approaches to prevent and/or treat bacterial infections. Differences between the siderophore-based iron uptake machineries of pathogenic Enterobacteriaceae and commensal microbes may lead to the development of selective "Trojan-horse" antimicrobials and immunization strategies that will not harm the host microbiota.
Collapse
Affiliation(s)
- Artur Sargun
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Romana R Gerner
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Manuela Raffatellu
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, University of California San Diego, La Jolla, California, USA.,Center for Microbiome Innovation, University of California San Diego, La Jolla, California, USA.,Chiba University-UC San Diego Center for Mucosal Immunology, Allergy, and Vaccines, La Jolla, California, USA
| | - Elizabeth M Nolan
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| |
Collapse
|