1
|
Li J, Chang J, Ma J, Zhou W, Yang Y, Wu J, Guan C, Yuan X, Xu L, Yu B, Su F, Ye S, Chen Y, Zhao G, Tang B. Genome-based assessment of antimicrobial resistance of Escherichia coli recovered from diseased swine in eastern China for a 12-year period. mBio 2025; 16:e0065125. [PMID: 40243369 PMCID: PMC12077178 DOI: 10.1128/mbio.00651-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Accepted: 03/24/2025] [Indexed: 04/18/2025] Open
Abstract
The global rise of antimicrobial resistance (AMR), driven by antibiotic use in healthcare and agriculture, poses a major public health threat. While AMR in clinical settings is well studied, there is a gap in understanding the resistance profiles of Escherichia coli from diseased livestock, particularly regarding zoonotic transmission. This study analyzes 114 E. coli isolates from diseased swine over 12 years, revealing that 99.12% were multidrug-resistant. Resistance was highest for ampicillin and amoxicillin/clavulanic acid (100%), followed by ciprofloxacin (96.49%) and tetracycline (94.74%). Furthermore, 21.05% of isolates were resistant to colistin, and 1.75% to tigecycline. A total of 76 antimicrobial resistance genes (ARGs) were identified, with mcr-1 found in 18.42%, mcr-3 in 4.39%, and tet(X4) in 1.75%. Significant co-occurrence of ARGs and plasmids suggests potential for co-selective dissemination. This study is the first to report enterotoxigenic E. coli (ETEC) strains carrying both mcr-1 and mcr-3 genes. After the 2017 colistin ban in China, mcr-1 detection rates significantly decreased, while florfenicol resistance rates increased in 2018-2021 (94.29%) compared to 2010-2017 (79.55%). This work provides valuable insights into the AMR profiles of E. coli from diseased swine and highlights trends that can inform strategies for monitoring and controlling public health risks associated with zoonotic E. coli transmission.IMPORTANCEThis study highlights the critical role of diseased and deceased swine in the spread of antimicrobial resistance (AMR), providing new insights into the transmission of resistance genes in zoonotic contexts. By analyzing E. coli from diseased swine, we identify key resistance genes such as mcr-1, mcr-3, and tet(X4), which pose significant public health risks, especially regarding last-resort antibiotics like colistin. Moreover, the study identifies novel transmission patterns of mcr genes, including ETEC strains carrying the mcr-3 gene and strains harboring both mcr-1 and mcr-3 genes. The role of plasmids in horizontal gene transfer is also revealed, facilitating rapid AMR spread across species. The long-term persistence of resistant strains highlights the challenges in controlling AMR in livestock. These findings underscore the need for enhanced surveillance and a One Health approach to mitigate AMR risks across animal, human, and environmental health.
Collapse
Affiliation(s)
- Junxing Li
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Jiang Chang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Jiangang Ma
- Xianghu Laboratory, Hangzhou, Zhejiang, China
| | - Wei Zhou
- Zhejiang Provincial Center for Animal Disease Prevention and Control, Hangzhou, China
| | - Yue Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Jing Wu
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Chunjiu Guan
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Xiufang Yuan
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Lihua Xu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Bin Yu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Fei Su
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Shiyi Ye
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Yijie Chen
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| | - Guoping Zhao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
- National Genomics Data Center & Bio-Med Big Data Center, CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Biao Tang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of the Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou, Zhejiang, China
| |
Collapse
|
2
|
Vickers TJ, Buckley DP, Khatoon N, Sheikh A, Setu B, Berndsen ZT, Fleckenstein JM. Parenteral vaccination with recombinant EtpA glycoprotein impairs enterotoxigenic E. coli colonization. Infect Immun 2025:e0060124. [PMID: 40310293 DOI: 10.1128/iai.00601-24] [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: 12/20/2024] [Accepted: 03/05/2025] [Indexed: 05/02/2025] Open
Abstract
Enterotoxigenic E. coli (ETEC) causes hundreds of millions of cases of acute diarrheal illness in low- and middle-income regions, disproportionately in young children. To date, there is no licensed, broadly protective vaccine against these common but antigenically heterogeneous pathogens. One of the more highly conserved antigens of ETEC, EtpA, is an extracellular glycoprotein adhesin that preferentially binds to A blood group glycans on intestinal epithelia. EtpA contributes to increased severity of illness in A blood group individuals, elicits robust serologic and fecal antibody responses following infection, and has been associated with protection against subsequent infection. However, its utility as a protective antigen needs further examination. In the present studies, we examined whether parenteral vaccination with recombinant EtpA (rEtpA) could afford protection against intestinal colonization in a murine model of ETEC infection. Here, we demonstrate that intramuscular vaccination with rEtpA, adjuvanted with double mutant LT (dmLT), primes IgG predominant mucosal antibody responses to ETEC challenge. Notably, however, both antibody levels and avidity, as well as protection, were dependent on the vaccination schedule. Likewise, through electron microscopy polyclonal epitope mapping (EMPEM), we observed a different repertoire of epitopes targeted by antibodies after a more protracted vaccination schedule. Next, we explored the utility of IM immunization with alum-adjuvanted rEtpA. This elicited strong serologic and fecal IgG responses. Although accompanied by negligible IgA mucosal responses, EtpA alum-adjuvanted IM vaccination nevertheless protected against ETEC intestinal colonization. Collectively, these data suggest that EtpA could expand the portfolio of antigens targeted in ETEC subunit vaccine development.
Collapse
Affiliation(s)
- Tim J Vickers
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - David P Buckley
- Department of Biochemistry, University of Missouri Columbia, Columbia, Missouri, USA
| | - Nazia Khatoon
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Alaullah Sheikh
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bipul Setu
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Zachary T Berndsen
- Department of Biochemistry, University of Missouri Columbia, Columbia, Missouri, USA
| | - James M Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- Infectious Diseases, Medicine Service, Veterans Affairs Saint Louis Health Care System, St. Louis, Missouri, USA
| |
Collapse
|
3
|
Kyu HH, Vongpradith A, Dominguez RMV, Ma J, Albertson SB, Novotney A, Khalil IA, Troeger CE, Doxey MC, Ledesma JR, Sirota SB, Bender RG, Swetschinski LR, Cunningham M, Spearman S, Abate YH, Abd Al Magied AHA, Abd ElHafeez S, Abdoun M, Abera B, Abidi H, Aboagye RG, Abtew YD, Abualruz H, Abu-Gharbieh E, Abukhadijah HJ, Aburuz S, Addo IY, Adekanmbi V, Adetunji COO, Adeyeoluwa TE, Adhikary RK, Adnani QES, Adra S, Adzigbli LA, Afolabi AA, Afzal MS, Afzal S, Agampodi SB, Agide FD, Ahinkorah BO, Ahmad A, Ahmad S, Ahmed A, Ahmed A, Ahmed H, Ahmed S, Akinosoglou K, Akter E, Al Awaidy S, Alajlani MM, Alam K, Albakri A, Albashtawy M, Aldhaleei WA, Algammal AM, Al-Gheethi AAS, Ali A, Ali SS, Ali W, Alif SM, Aljunid SM, Al-Marwani S, Almazan JU, Al-Mekhlafi HM, Almustanyir S, Alqahatni SA, Alrawashdeh A, Al-Rifai RH, Alsabri MA, Altaf A, Altirkawi KA, Alvis-Guzman N, Alvis-Zakzuk NJ, Alyahya MSI, Al-Zyoud WA, Amugsi DA, Andrei CL, Antoni S, Anuoluwa BS, Anuoluwa IA, Anwar S, Anwari P, Apostol GLC, Arabloo J, Arafat M, Aravkin AY, Areda D, Aregawi BB, Aremu A, Arndt MB, Asgedom AA, Ashraf T, Athari SS, Atreya A, Ayele F, Azadi D, Azhar GS, Aziz S, Azzam AY, et alKyu HH, Vongpradith A, Dominguez RMV, Ma J, Albertson SB, Novotney A, Khalil IA, Troeger CE, Doxey MC, Ledesma JR, Sirota SB, Bender RG, Swetschinski LR, Cunningham M, Spearman S, Abate YH, Abd Al Magied AHA, Abd ElHafeez S, Abdoun M, Abera B, Abidi H, Aboagye RG, Abtew YD, Abualruz H, Abu-Gharbieh E, Abukhadijah HJ, Aburuz S, Addo IY, Adekanmbi V, Adetunji COO, Adeyeoluwa TE, Adhikary RK, Adnani QES, Adra S, Adzigbli LA, Afolabi AA, Afzal MS, Afzal S, Agampodi SB, Agide FD, Ahinkorah BO, Ahmad A, Ahmad S, Ahmed A, Ahmed A, Ahmed H, Ahmed S, Akinosoglou K, Akter E, Al Awaidy S, Alajlani MM, Alam K, Albakri A, Albashtawy M, Aldhaleei WA, Algammal AM, Al-Gheethi AAS, Ali A, Ali SS, Ali W, Alif SM, Aljunid SM, Al-Marwani S, Almazan JU, Al-Mekhlafi HM, Almustanyir S, Alqahatni SA, Alrawashdeh A, Al-Rifai RH, Alsabri MA, Altaf A, Altirkawi KA, Alvis-Guzman N, Alvis-Zakzuk NJ, Alyahya MSI, Al-Zyoud WA, Amugsi DA, Andrei CL, Antoni S, Anuoluwa BS, Anuoluwa IA, Anwar S, Anwari P, Apostol GLC, Arabloo J, Arafat M, Aravkin AY, Areda D, Aregawi BB, Aremu A, Arndt MB, Asgedom AA, Ashraf T, Athari SS, Atreya A, Ayele F, Azadi D, Azhar GS, Aziz S, Azzam AY, Babu GR, Bahrami Taghanaki P, Bahramian S, Balakrishnan S, Banik B, Bante SA, Bardhan M, Bärnighausen TW, Barqawi HJ, Barrow A, Basharat Z, Bassat Q, Bastan MM, Basu S, Bathini PP, Behzadi P, Beiranvand M, Bello MB, Bello OO, Beloukas A, Beran A, Bhandari D, Bhardwaj P, Bhutta ZA, Borhany H, Bouaoud S, Brauer M, Buonsenso D, Butt ZA, Çakmak Barsbay M, Cámera LA, Capodici A, Castañeda-Orjuela CA, Cenderadewi M, Chakraborty C, Chakraborty S, Chattu VK, Chaudhary AA, Chichagi F, Ching PR, Chirinos-Caceres JL, Chopra H, Choudhari SG, Chowdhury EK, Chu DT, Chukwu IS, Chutiyami M, Cruz-Martins N, Dadras O, Dai X, Dandona L, Dandona R, Darcho SD, Das JK, Dash NR, Delgado-Enciso I, Desye B, Devanbu VGC, Dhama K, Dhimal M, Diaz MJ, Do TC, Dohare S, Dorostkar F, Doshi OP, Doshmangir L, Dsouza HL, Duraisamy S, Durojaiye OC, E'mar AR, Ed-Dra A, Edinur HA, Efendi D, Efendi F, Eghbali F, Ekundayo TC, El Sayed I, Elhadi M, El-Metwally AA, Elshaer M, Elsohaby I, Eltaha C, Eshrati B, Eslami M, Fahim A, Fakhradiyev IR, Fakhri-Demeshghieh A, Farahmand M, Fasina FO, Fasina MM, Feizkhah A, Fekadu G, Ferreira N, Fetensa G, Fischer F, Fukumoto T, Fux B, Gadanya MA, Gaihre S, Gajdács M, Galali Y, Gandhi AP, Gautam RK, Gebregergis MW, Gebrehiwot M, Gebremeskel TG, Getachew ME, Getahun GK, Getie M, Ghasemzadeh A, Ghazy RM, Ghozy S, Gil AU, Girmay AA, Gizaw ATT, Golechha M, Goleij P, Gona PN, Grada A, Guarducci G, Gudeta MD, Gupta VK, Habteyohannes AD, Hadi NR, Hamidi S, Hamilton EB, Harapan H, Hasan MK, Hasan SM, Hasani H, Hasnain MS, Hassan II, He J, Hemmati M, Hezam K, Hosseinzadeh M, Huang J, Huynh HH, Ibitoye SE, Ikuta KS, Ilesanmi OS, Ilic IM, Ilic MD, Inamdar S, Isa MA, Islam MR, Islam SMS, Ismail NE, Iwu CD, Jacobsen KH, Jahrami H, Jain A, Jain N, Jairoun AA, Jakovljevic M, Jalilzadeh Yengejeh R, Javidnia J, Jayaram S, Jokar M, Jonas JB, Joseph A, Joseph N, Jozwiak JJ, Kabir H, Kadir DHH, Kamal MM, Kamal VK, Kamireddy A, Kanchan T, Kanmodi KK, Kannan S S, Kantar RS, Karami J, Karki P, Kasraei H, Kaur H, Keykhaei M, Khader YS, Khalilian A, Khamesipour F, Khan G, Khan MJ, Khan ZA, Khanal V, Khatab K, Khatatbeh MM, Khater AM, Kheirallah KA, Khidri FF, Khosla AA, Kim K, Kim YJ, Kisa A, Kissoon N, Klu D, Kochhar S, Kolahi AA, Kompani F, Kosen S, Krishan K, Kuate Defo B, Kuddus MA, Kuddus M, Kulimbet M, Kumar GA, Kumar R, Kyei-Arthur F, Lahariya C, Lal DK, Le NHH, Lee SW, Lee WC, Lee YY, Li MC, Ligade VS, Liu G, Liu S, Liu X, Liu X, Lo CH, Lucchetti G, Lv L, Malhotra K, Malik AA, Marasini BP, Martorell M, Marzo RR, Masoumi-Asl H, Mathur M, Mathur N, Mediratta RP, Meftah E, Mekene Meto T, Meles HN, Melese EB, Mendoza W, Merati M, Meretoja TJ, Mestrovic T, Mettananda S, Minh LHN, Mishra V, Mithra P, Mohamadkhani A, Mohamed AI, Mohamed MFH, Mohamed NS, Mohammed M, Mohammed S, Monasta L, Moni MA, Motappa R, Mougin V, Mubarik S, Mulita F, Munjal K, Munkhsaikhan Y, Naghavi P, Naik G, Nair TS, Najmuldeen HHR, Nargus S, Narimani Davani D, Nashwan AJ, Natto ZS, Nazri-Panjaki A, Nchanji GT, Ndishimye P, Ngunjiri JW, Nguyen DH, Nguyen NNY, Nguyen VT, Nigatu YT, Nikoobar A, Niranjan V, Nnaji CA, Noman EA, Noor NM, Noor STA, Nouri M, Nozari M, Nri-Ezedi CA, Nugen F, Odetokun IA, Ogunfowokan AA, Ojo-Akosile TR, Okeke IN, Okekunle AP, Olorukooba AA, Olufadewa II, Oluwatunase GO, Orish VN, Ortega-Altamirano DV, Ortiz-Prado E, Osuagwu UL, Osuolale O, Ouyahia A, Padubidri JR, Pandey A, Pandey A, Pando-Robles V, Pardhan S, Parikh RR, Patel J, Patil S, Pawar S, Peprah P, Perianayagam A, Perna S, Petcu IR, Philip AK, Polibin RV, Postma MJ, Pourtaheri N, Pradhan J, Prates EJS, Pribadi DRA, Qasim NH, Qazi AS, R D, Radhakrishnan V, Rahim F, Rahman M, Rahman MA, Rahmani S, Rahmanian M, Rahmanian N, Ramadan MM, Ramasamy SK, Ramazanu S, Rameto MAA, Ramteke PW, Rana K, Ranabhat CL, Rasella D, Rashidi MM, Rasouli-Saravani A, Rathish D, Rauniyar SK, Rawaf S, Redwan EMM, Regmi AR, Rengasamy KRR, Rezaei N, Rezaei N, Rezaeian M, Riad A, Rodrigues M, Rodriguez JAB, Roever L, Rohilla R, Ronfani L, Rony MKK, Ross AG, Roudashti S, Roy B, Runghien T, Sachdeva Dhingra M, Saddik BA, Sadeghi E, Safari M, Sahoo SS, Sajadi SM, Salami AA, Saleh MA, Samadi Kafil H, Samodra YL, Sanabria J, Sanjeev RK, Sarkar T, Sartorius B, Sathian B, Satpathy M, Sawhney M, Schumacher AE, Sebsibe MA, Serban D, Shafie M, Shahid S, Shahid W, Shaikh MA, Sham S, Shamim MA, Shams-Beyranvand M, Shamshirgaran MA, Shanawaz M, Shannawaz M, Sharifan A, Sharma M, Sharma V, Shenoy SM, Sherchan SP, Shetty M, Shetty PH, Shiferaw D, Shittu A, Shorofi SA, Siddig EE, Silva LMLR, Singh B, Singh JA, Sinto R, Socea B, Soeters HM, Sokhan A, Sood P, Soraneh S, Sreeramareddy CT, Srinivasamurthy SK, Srivastava VK, Stanikzai MH, Subedi N, Subramaniyan V, Sulaiman SK, Suleman M, Swain CK, Szarpak L, T Y SS, Tabatabaei SM, Tabche C, Taha ZMA, Talukder A, Tamuzi JL, Tan KK, Tandukar S, Temsah MH, Thakali O, Thakur R, Thirunavukkarasu S, Thomas J, Thomas NK, Ticoalu JHV, Tiwari K, Tovani-Palone MR, Tram KH, Tran AT, Tran NM, Tran TH, Tromans SJ, Truyen TTTT, Tumurkhuu M, Udoakang AJ, Udoh A, Ullah S, Umair M, Umar M, Unim B, Unnikrishnan B, Vahdati S, Vaithinathan AG, Valizadeh R, Verma M, Verras GI, Vinayak M, Waheed Y, Walde MT, Wang Y, Waqas M, Weerakoon KG, Wickramasinghe ND, Wolde AA, Wu F, Yaghoubi S, Yaya S, Yezli S, Yiğit V, Yin D, Yon DK, Yonemoto N, Yusuf H, Zahid MH, Zakham F, Zaki L, Zare I, Zastrozhin M, Zeariya MGM, Zhang H, Zhang ZJ, Zhumagaliuly A, Zia H, Zoladl M, Mokdad AH, Lim SS, Vos T, Platts-Mills JA, Mosser JF, Reiner RC, Hay SI, Naghavi M, Murray CJL. Global, regional, and national age-sex-specific burden of diarrhoeal diseases, their risk factors, and aetiologies, 1990-2021, for 204 countries and territories: a systematic analysis for the Global Burden of Disease Study 2021. THE LANCET. INFECTIOUS DISEASES 2025; 25:519-536. [PMID: 39708822 PMCID: PMC12018300 DOI: 10.1016/s1473-3099(24)00691-1] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/09/2024] [Accepted: 10/14/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND Diarrhoeal diseases claim more than 1 million lives annually and are a leading cause of death in children younger than 5 years. Comprehensive global estimates of the diarrhoeal disease burden for specific age groups of children younger than 5 years are scarce, and the burden in children older than 5 years and in adults is also understudied. We used results from the Global Burden of Diseases, Injuries, and Risk Factors Study 2021 to assess the burden of, and trends in, diarrhoeal diseases overall and attributable to 13 pathogens, as well as the contributions of associated risk factors, in children and adults in 204 countries and territories from 1990 to 2021. METHODS We used the Cause of Death Ensemble modelling strategy to analyse vital registration data, verbal autopsy data, mortality surveillance data, and minimally invasive tissue sampling data. We used DisMod-MR (version 2.1), a Bayesian meta-regression tool, to analyse incidence and prevalence data identified via systematic reviews, population-based surveys, and claims and inpatient data. We calculated diarrhoeal disability-adjusted life-years (DALYs) as the sum of years of life lost (YLLs) and years lived with disability (YLDs) for each location, year, and age-sex group. For aetiology estimation, we used a counterfactual approach to quantify population-attributable fractions (PAFs). Additionally, we estimated the diarrhoeal disease burden attributable to the independent effects of risk factors using the comparative risk assessment framework. FINDINGS In 2021, diarrhoeal diseases caused an estimated 1·17 million (95% uncertainty interval 0·793-1·62) deaths globally, representing a 60·3% (50·6-69·0) decrease since 1990 (2·93 million [2·31-3·73] deaths). The most pronounced decline was in children younger than 5 years, with a 79·2% (72·4-84·6) decrease in diarrhoeal deaths. Global YLLs also decreased substantially, from 186 million (147-221) in 1990 to 51·4 million (39·9-65·9) in 2021. In 2021, an estimated 59·0 million (47·2-73·2) DALYs were attributable to diarrhoeal diseases globally, with 30·9 million (23·1-42·0) of these affecting children younger than 5 years. Leading risk factors for diarrhoeal DALYs included low birthweight and short gestation in the neonatal age groups, child growth failure in children aged between 1-5 months and 2-4 years, and unsafe water and poor sanitation in older children and adults. We estimated that the removal of all evaluated diarrhoeal risk factors would reduce global DALYs from 59·0 million (47·2-73·2) to 4·99 million (1·99-10·0) among all ages combined. Globally in 2021, rotavirus was the predominant cause of diarrhoeal deaths across all ages, with a PAF of 15·2% (11·4-20·1), followed by norovirus at 10·6% (2·3-17·0) and Cryptosporidium spp at 10·2% (7·03-14·3). In children younger than 5 years, the fatal PAF of rotavirus was 35·2% (28·7-43·0), followed by Shigella spp at 24·0% (15·2-37·9) and adenovirus at 23·8% (14·8-36·3). Other pathogens with a fatal PAF greater than 10% in children younger than 5 years included Cryptosporidium spp, typical enteropathogenicEscherichia coli, and enterotoxigenic E coli producing heat-stable toxin. INTERPRETATION The substantial decline in the global burden of diarrhoeal diseases since 1990, particularly in children younger than 5 years, supports the effectiveness of health interventions such as oral rehydration therapy, enhanced water, sanitation, and hygiene (WASH) infrastructure, and the introduction and scale-up of rotavirus vaccination. Targeted interventions and preventive measures against key risk factors and pathogens could further reduce this burden. Continued investment in the development and distribution of vaccines for leading pathogens remains crucial. FUNDING Bill & Melinda Gates Foundation.
Collapse
|
4
|
Chen J, Jia X, Hu Y, Zhao X, Cheng Y, Lu L, Zhong S, You J, Zou T. Benzoic acid as a dietary supplement mitigates inflammation and intestinal injury in acute enterotoxigenic Escherichia coli-infected mice without adverse effects in healthy mice. Food Funct 2025; 16:3195-3210. [PMID: 40190113 DOI: 10.1039/d5fo00514k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Benzoic acid is a naturally occurring compound found in fruits and is also commercially synthesized as an additive in the food, feed, and pharmaceutical industries. This study investigated the effects of benzoic acid as a dietary supplement on inflammation and intestinal injury in acute Escherichia coli (ETEC)-infected or healthy mice. Thirty-six BALB/c mice were divided into three groups, with 12 mice in each group for a 16-day feeding trial. In group 1, mice were fed a basal diet, six mice were sacrificed, and six mice were intraperitoneally injected with phosphate-buffered saline on day 15. Groups 2 and 3 were fed a basal diet and a diet containing 0.6% benzoic acid, respectively. Half of the mice in each group were sacrificed, while the others were intraperitoneally injected with ETEC on day 15. The results indicated that benzoic acid had no adverse effects on healthy mice regarding growth, organ indices, inflammation, intestinal injury parameters, and cecal short-chain fatty acid levels. Importantly, benzoic acid reduced inflammation in ETEC-infected mice, as evidenced by decreased serum IL-1β, TNF-α, and INF-γ levels, along with increased jejunal TLR-2 and MyD88 mRNA expression. Besides, benzoic acid mitigated intestinal injury in ETEC-infected mice by increasing the jejunal villus height (VH) and the ratio of VH to crypt depth, elevating jejunal Occludin mRNA levels, decreasing serum D-lactate and diamine oxidase levels, and increasing the cecal acetic acid level. 16s rRNA sequencing revealed that benzoic acid altered the β-diversity of ETEC-infected mice and increased the abundances of Erysipelotrichaceae, Faecalibaculum, and Turicibacter in their gut microbiota. Spearman correlation analysis further indicated that the protective effects of benzoic acid against ETEC infection were closely linked to specific gut microbiota, namely Erysipelotrichaceae, Faecalibaculum, Bifidobacterium, and Limosilactobacillus. Collectively, these findings suggest that benzoic acid could serve as a safe dietary supplement for healthy mice and may alleviate inflammation and intestinal injury in mice with acute ETEC infection.
Collapse
Affiliation(s)
- Jun Chen
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Xuena Jia
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Youjun Hu
- Guangdong Nuacid Nutrition Co., Ltd, Qingyuan 511500, China
| | - Xiaonan Zhao
- Guangdong Nuacid Nutrition Co., Ltd, Qingyuan 511500, China
| | - Yong Cheng
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Li Lu
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Songtao Zhong
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Jinming You
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China.
| | - Tiande Zou
- Jiangxi Province Key Laboratory of Animal Nutrition and Feed, Jiangxi Province Key Innovation Center of Integration in Production and Education for High-quality and Safe Livestock and Poultry, Jiangxi Agricultural University, Nanchang 330045, China.
| |
Collapse
|
5
|
Sthity RA, Islam MZ, Sagar MEK, Gazi MA, Ferdous J, Kabir MM, Mahfuz M, Ahmed T, Mostafa I. Association of Escherichia coli pathotypes with fecal markers of enteropathy and nutritional status among underweight adults in Bangladesh. Front Cell Infect Microbiol 2025; 15:1553688. [PMID: 40276386 PMCID: PMC12018316 DOI: 10.3389/fcimb.2025.1553688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Introduction Environmental enteric dysfunction (EED), a subclinical intestinal disorder, is characterized by chronic fecal-oral exposure to entero-pathogens and could be diagnosed by measuring non-invasive biomarkers. Escherichia coli is the one of the key bacterial enteric pathogens that drives EED, but there is a lack of information on the E. coli pathotypes in relation to the biomarkers of EED in malnourished adults. Here, we intended to measure the possible association of these pathotypes with EED biomarkers and nutritional status of adults residing in a slum in Bangladesh. Method Fecal samples were collected from 524 malnourished adults (BMI ≤18.5 kg/m2) living in a slum-setting in Dhaka from March 2016 to September 2019 and analyzed by TaqMan Array Card assays to evaluate the presence of E. coli pathotypes and other entero-pathogens. The multivariable linear regression model was used to assess the association. Results In these malnourished adults, the most prevalent pathotype of E. coli was EAEC (61.7%) and the least prevalent was STEC (6.7%). The prevalence of atypical EPEC, ETEC and Shigella/EIEC were 52%, 48.9% and 45.1% respectively. The infection with atypical EPEC had significant positive association with levels of Myeloperoxidase (b = 0.38; 95% CI = 0.11, 0.65; p-value = 0.006). Similarly, a significantly higher concentration of alpha-1-antitrypsin (b = 0.13; 95% CI = 0.03, 0.22; p-value = 0.011) was found in the STEC-infected adults. However, no notable association was found between the E. coli pathotypes and nutritional status of these adult participants. Moreover, Plesiomonas infected adults were more likely to be infected with EAEC (p-value = 0.017), ETEC (p-value <0.001) and STEC (pvalue = 0.002). Significant coinfection was also detected among the pathotypes and other entero-pathogens such as Giardia, Ascaris, Campylobacter, Salmonella, Enterocytozoon bieneusi, and Adenovirus. Discussion The study results imply that there is an influence of particular E. coli pathotypes (EPEC and STEC) on intestinal inflammation and gut permeability of the malnourished Bangladeshi adults, but no association with nutritional status is found. Potential pathogenicity of the E. coli pathotypes is also observed when co-infection with other pathogens exists in these adults.
Collapse
Affiliation(s)
- Rahvia Alam Sthity
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Zahidul Islam
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Ehsanul Kabir Sagar
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Amran Gazi
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, United States
| | - Jafrin Ferdous
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Md. Mamun Kabir
- Infectious Disease Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Mustafa Mahfuz
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Tahmeed Ahmed
- Office of the Executive Director, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Department of Public Health Nutrition, James P Grant School of Public Health, BRAC University, Dhaka, Bangladesh
| | - Ishita Mostafa
- Nutrition Research Division, International Centre for Diarrhoeal Disease Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
6
|
Walker RI. Conserved antigens for enteric vaccines. Vaccine 2025; 50:126828. [PMID: 39914256 PMCID: PMC11878282 DOI: 10.1016/j.vaccine.2025.126828] [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: 10/01/2024] [Revised: 01/23/2025] [Accepted: 01/28/2025] [Indexed: 02/25/2025]
Abstract
Enterotoxigenic Escherichia coli (ETEC), Shigella, and Campylobacter have been identified as major causes of diarrheal diseases worldwide. In addition to overt disease and death, they are responsible for stunting in children with the risk of lifelong consequences on health and economic opportunities. All three of these bacterial pathogens, which collectively account for approximately 30 % of the cases of diarrheal diseases, are recognized as antimicrobial resistance (AMR) threats. In spite of the dangers these pathogens represent for both children and adults, there is as yet no licensed vaccine available for any of them. Fortunately, much has been accomplished to identify conserved antigens against each of these pathogens so that now relatively simple vaccines have the potential to be developed into multi-pathogen vaccines which could have a major impact on reduction of diarrheal diseases. Conserved antigens may be used even more efficiently if consolidated and expressed on a cellular vector or as part of a conjugate vaccine. A new mucosal adjuvant, double mutant heat-labile toxin (dmLT), has been shown to not only be among the conserved antigens against ETEC, but to also have properties which drive robust mucosal and systemic immune responses for antigens given orally or intramuscularly. Conserved antigens and the strategies for their use such as co-administration with dmLT will be presented in this review.
Collapse
Affiliation(s)
- Richard I Walker
- PATH, 455 Massachusetts Ave, Suite 1000, Washington, DC, 20001-2621, USA.
| |
Collapse
|
7
|
Liu J, Jokiranta TS, Carlin N, Stroup S, Zhang J, Sjostrand B, Svennerholm AM, Houpt ER, Kantele A. Use of a TaqMan Array Card for identification of enterotoxins and colonization factors directly from stool samples in an enterotoxigenic E. coli vaccine study. Microbiol Spectr 2025; 13:e0187024. [PMID: 39932427 PMCID: PMC11878035 DOI: 10.1128/spectrum.01870-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 01/06/2025] [Indexed: 03/05/2025] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a leading cause of childhood and travelers' diarrhea. The vaccine candidate ETVAX encompasses several ETEC colonization factors (CFs) with a hybrid LT (heat-labile toxin)/cholera toxin B subunit adjuvanted with a double-mutant LT. Stool samples from a Phase 2b ETVAX trial were tested by a PCR-based customized TaqMan Array Card (TAC), including three ETEC toxin genes (LT and heat-stable toxins, STh and STp) and 18 ETEC CFs. Stool samples were also tested with the molecular platform Amplidiag and culture, followed by GM1-enzyme-linked immunosorbent assay (ELISA) and inhibition GM1-ELISA for LT and ST and dot blot for CFs of ETECs identified among six culture isolates (maximum). Compared with Amplidiag, TAC yielded 89.4% sensitivity (320/358) and 96.4% specificity (405/420) for ETEC detection. The two methods demonstrated a good quantitative correlation (quantification cycle R2 = 0.827, P < 0.05). Compared with culture, TAC and Amplidiag each exhibited 96.8% (184/190) sensitivity and identified an additional of 151 and 174 PCR positives in 588 culture-negative stools, respectively. The concordance of stool TAC versus ELISA of ETEC colonies for LT and STh/STp was 85.5% (165/193). TAC demonstrated 98% sensitivity and 92% specificity versus the dot blot results of 793 colonies for the ETVAX CFs CFA/I, CS3, CS5, and CS6. Overall ETEC was detected by TAC in 335 (43.1%) and by Amplidiag in 358 (46.0%) of specimens compared to 190 (24.4%) by culture. We conclude that molecular diagnostic approaches of TAC or Amplidiag increase the detection of ETEC compared with culture, and TAC can also provide vaccine-subtype ETEC data.CLINICAL TRIALSThis study was registered with ClinicalTrials.gov as NCT03729219.IMPORTANCEEnterotoxigenic Escherichia coli (ETEC) is an important cause of childhood and travelers' diarrhea. Vaccines in development utilize specific toxins and colonization factors (CFs) as antigens. Therefore, clinical microbiologic diagnostic methods are needed to discriminate specific toxins and CFs, both for vaccine trials and to guide epidemiology. In this work, we assessed the diagnostic performance of several methods for ETEC: a PCR-based customized TaqMan Array Card (TAC) and the molecular platform Amplidiag on stool and E. coli culture, followed by GM1-enzyme-linked immunosorbent assay for toxins and dot blot for CFs. Stool samples from a Phase 2b ETEC vaccine trial were used. Overall, ETEC was detected by TAC in 335 (43.1%) and by Amplidiag in 358 samples (46.0%) compared to 190 (24.4%) by culture. TAC additionally provided CF data with 98% sensitivity and 92% specificity. We conclude that the molecular diagnostic approaches of TAC or Amplidiag increase the detection of ETEC compared with culture.
Collapse
Affiliation(s)
- Jie Liu
- School of Public Health, Qingdao University, Qingdao, China
| | - T. Sakari Jokiranta
- Department of Bacteriology and Immunology, Medicum, University of Helsinki, Helsinki, Finland
| | | | - Suzanne Stroup
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Jixian Zhang
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | | | - Ann-Mari Svennerholm
- Gothenburg University Vaccine Research Institute, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Eric R. Houpt
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, Virginia, USA
| | - Anu Kantele
- Meilahti Vaccine Research Center MeVac, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
8
|
Salvador-Erro J, Pastor Y, Gamazo C. Targeting Enterotoxins: Advancing Vaccine Development for Enterotoxigenic Escherichia coli ETEC. Toxins (Basel) 2025; 17:71. [PMID: 39998088 PMCID: PMC11860656 DOI: 10.3390/toxins17020071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a major cause of diarrheal disease worldwide, particularly in children in low- and middle-income countries. Its ability to rapidly colonize the intestinal tract through diverse colonization factors and toxins underpins its significant public health impact. Despite extensive research and several vaccine candidates reaching clinical trials, no licensed vaccine exists for ETEC. This review explores the temporal and spatial coordination of ETEC virulence factors, focusing on the interplay between adherence mechanisms and toxin production as critical targets for therapeutic intervention. Advancements in molecular biology and host-pathogen interaction studies have uncovered species-specific variations and cross-reactivity between human and animal strains. In particular, the heat-labile (LT) and heat-stable (ST) toxins have provided crucial insights into molecular mechanisms and intestinal disruption. Additional exotoxins, such as EAST-1 and hemolysins, further highlight the multifactorial nature of ETEC pathogenicity. Innovative vaccine strategies, including multiepitope fusion antigens (MEFAs), mRNA-based approaches, and glycoconjugates, aim to enhance broad-spectrum immunity. Novel delivery methods, like intradermal immunization, show promise in eliciting robust immune responses. Successful vaccination against ETEC will offer an effective and affordable solution with the potential to greatly reduce mortality and prevent stunting, representing a highly impactful and cost-efficient solution to a critical global health challenge.
Collapse
Affiliation(s)
| | | | - Carlos Gamazo
- Department of Microbiology and Parasitology, Navarra Medical Research Institute (IdiSNA), University of Navarra, 31008 Pamplona, Spain; (J.S.-E.); (Y.P.)
| |
Collapse
|
9
|
Pasetti MF, Milletich PL, White JA, Butts J, Brady RC, Dickey MD, Ballou C, Maier N, Sztein MB, Baqar S, Louis Bourgeois A, Bernstein DI. Safety and immunogenicity in humans of enterotoxigenic Escherichia coli double mutant heat-labile toxin administered intradermally. NPJ Vaccines 2025; 10:23. [PMID: 39893179 PMCID: PMC11787345 DOI: 10.1038/s41541-025-01071-7] [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: 10/04/2024] [Accepted: 01/09/2025] [Indexed: 02/04/2025] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) diarrhea is associated with a high burden of disease globally, for which no licensed vaccine is available. A Phase 1, double-blind, dose-escalation (0.1-2.0 µg) study was conducted to evaluate the safety and immunogenicity of double mutant heat-labile toxin LTR192G/L211A (dmLT) delivered intradermally (ID) to healthy adults. Subjects received up to three immunizations at three-week intervals. The vaccine was safe, although it induced mild local and some gastrointestinal adverse events, as well as frequent hyperpigmentation at the injection site. High levels of serum IgG and IgA, LT neutralizing antibodies, and IgG and IgA antibodies in lymphocyte supernatant were elicited post-vaccination, most prominently at the largest dose (2.0 μg). Rates of responses were the highest in subjects who received the largest dose (2.0 μg) and multiple immunizations. The ETEC dmLT vaccine was safe and highly immunogenic, inducing long-lasting systemic and mucosal responses when administered by the ID route. Trial registration Clinical Trials NCT02531685.
Collapse
Affiliation(s)
- Marcela F Pasetti
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Patricia L Milletich
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | - Rebecca C Brady
- Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michelle D Dickey
- Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Nicole Maier
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Marcelo B Sztein
- Center for Vaccine Development and Global Health and Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Shahida Baqar
- Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - A Louis Bourgeois
- PATH, Washington, DC, USA
- John Hopkins University School of Public Health, Baltimore, MD, USA
| | - David I Bernstein
- Cincinnati Children's Hospital Medical Center; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
10
|
Kuijper EJ, Gerding DN. The End of Toxoid Vaccine Development for Preventing Clostridioides difficile Infections? Clin Infect Dis 2024; 79:1512-1514. [PMID: 39178347 DOI: 10.1093/cid/ciae412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/10/2024] [Accepted: 08/21/2024] [Indexed: 08/25/2024] Open
Abstract
Abstract
Collapse
Affiliation(s)
- Ed J Kuijper
- National Expertise Center for Clostridioides difficile Infections of Leiden University Center for Infectious Diseases, Leiden University Medical Centre (LUMC), Leiden, The Netherlands and Center for Infectious Disease Control (CIb), National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Dale N Gerding
- Edward Hines, Jr, Veterans Affairs Hospital, Hines, Illinois, USA
| |
Collapse
|
11
|
Riaz S, Steinsland H, Andersen AZ, Boysen A, Hanevik K. Proportions of IgA antibodies targeting glycosylated epitopes of secreted Escherichia coli mucinase YghJ in initial plasmablast response differ from salivary and intestinally secreted IgA. Med Microbiol Immunol 2024; 214:2. [PMID: 39673573 DOI: 10.1007/s00430-024-00812-0] [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: 07/15/2024] [Accepted: 11/24/2024] [Indexed: 12/16/2024]
Abstract
Mucosal infections normally cause an immune response including activation of antigen-specific B cells in regional mucosa-associated lymphoid tissue. After recirculation of plasmablasts, and maturation at mucosal surfaces or bone marrow, plasma cells produce secretory or systemic IgA. It remains uncertain to what extent secretory and systemic IgA share the same target specificities. For vaccine candidate optimization, it is important to know whether IgA targeting of glycosylated epitopes of a protein antigen vary between mucosal and systemic sites. We evaluated glycosylated epitope specificity of systemic and mucosally secreted IgA against YghJ, a potential vaccine candidate antigen secreted by most pathogenic Escherichia coli. IgA from intestinal lavage, saliva, serum, and blood-derived antibody in lymphocyte supernatants (ALS) were collected from 21 volunteers following experimental infection with enterotoxigenic E. coli. Methods for preparing IgA from saliva and ALS were developed, and multiplex bead flow cytometric immunoassays were used to determine levels of IgA targeting natively glycosylated YghJ and estimating what proportion of these antibodies specifically targeted glycosylated epitopes. Following infection, anti-YghJ IgA levels increased substantially for most volunteers across all four specimen types. Target specificity of ALS IgA correlated well with serum IgA, but not with mucosally secreted IgA. Furthermore, glycosylation-specific proportion of salivary IgA was higher than, and did not correlate with, intestinally secreted IgA. These results indicate a new degree of complexity to our understanding of epitope-targeting and tissue specificity of mucosal antibody responses. Our findings also suggest that all features of an intestinal IgA response may not be well reflected in serum, saliva, or ALS, which are commonly used proxy specimens for evaluating intestinal immune responses.
Collapse
Affiliation(s)
- Saman Riaz
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Hans Steinsland
- Centre for Intervention Science in Maternal and Child Health (CISMAC), Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | | | | | - Kurt Hanevik
- Department of Clinical Science, University of Bergen, Bergen, Norway.
- National Centre for Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
12
|
Bao X, Wu J. Natural anti-adhesive components against pathogenic bacterial adhesion and infection in gastrointestinal tract: case studies of Helicobacter pylori, Salmonella enterica, Clostridium difficile, and diarrheagenic Escherichia coli. Crit Rev Food Sci Nutr 2024:1-46. [PMID: 39666022 DOI: 10.1080/10408398.2024.2436139] [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
Antimicrobial resistance (AMR) poses a global public health concern. Recognizing the critical role of bacterial adhesion in pathogenesis of infection, anti-adhesive therapy emerges as a promising approach to impede initial bacterial attachment, thus preventing pathogenic colonization and infection. Natural anti-adhesive agents derived from food sources are generally safe and have the potential to inhibit the emergence of resistant bacteria. This comprehensive review explored diverse natural dietary components exhibiting anti-adhesive activities against several model enteric pathogens, including Helicobacter pylori, Salmonella enterica, Clostridium difficile, and three key diarrheagenic Escherichia coli (i.e., enterotoxigenic E. coli, enteropathogenic E. coli, and enterohemorrhagic E. coli). Investigating various anti-adhesive products will advance our understanding of current research of the field and inspire further development of these agents as potential nutraceuticals or adjuvants to improve the efficacy of conventional antibiotics.
Collapse
Affiliation(s)
- Xiaoyu Bao
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| | - Jianping Wu
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
13
|
Watanabe-Takahashi M, Tanigawa T, Hamabata T, Nishikawa K. A tetravalent peptide efficiently inhibits the intestinal toxicity of heat-labile enterotoxin by targeting the receptor-binding region of the B-subunit pentamer. Biochem Biophys Res Commun 2024; 734:150769. [PMID: 39369542 DOI: 10.1016/j.bbrc.2024.150769] [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: 09/08/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/08/2024]
Abstract
Infection by enterotoxigenic Escherichia coli (ETEC) causes severe watery diarrhea and dehydration in humans. Heat-labile enterotoxin (LT) is a major virulence factor produced by ETEC. LT is one of AB5-type toxins, such as Shiga toxin (Stx) and cholera toxin (Ctx), and the B-subunit pentamer is responsible for high affinity binding to the LT-receptor, ganglioside GM1, through multivalent interaction. In this report, we found that Glu51 of the B-subunit plays an essential role in receptor binding compared with other amino acids, such as Glu11, Arg13, and Lys91, all of which were previously shown to be involved in the binding. By targeting Glu51, we identified four tetravalent peptides that specifically bind to the B-subunit pentamer with high affinity by screening tetravalent random-peptide libraries, which were tailored to bind to the B-subunit through multivalent interaction. One of these peptides, GGR-tet, efficiently inhibited the cell-elongation phenotype and the elevation of cellular cAMP levels, both induced by LT. Furthermore, GGR-tet markedly inhibited LT-induced fluid accumulation in the mouse ileum. Thus, GGR-tet represents a novel therapeutic agent against ETEC infection.
Collapse
Affiliation(s)
- Miho Watanabe-Takahashi
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, Japan.
| | - Tetsuya Tanigawa
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, Japan
| | - Takashi Hamabata
- Department of Infectious Disease, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kiyotaka Nishikawa
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, Japan.
| |
Collapse
|
14
|
Ashton PM, Katuah ZJ, Botomani A, Kutambe BM, Cunliffe NA, von Mentzer A, Msefula C, Jere KC. Enterotoxigenic Escherichia coli in Blantyre, Malawi. Access Microbiol 2024; 6:000885.v3. [PMID: 39559263 PMCID: PMC11572489 DOI: 10.1099/acmi.0.000885.v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 09/09/2024] [Indexed: 11/20/2024] Open
Abstract
We announce the deposition of the first two enterotoxigenic Escherichia coli (ETEC) genomes from Malawi. They were isolated from the faeces of asymptomatically infected children obtained in 2014. Both genomes encode the porcine variant of the heat-labile toxin and no known ETEC colonization factors.
Collapse
Affiliation(s)
- Philip M. Ashton
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi
- Institute of Infection, Veterinary, and Ecological Sciences, University of Liverpool, Liverpool, UK
| | | | | | | | - Nigel A. Cunliffe
- Institute of Infection, Veterinary, and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Astrid von Mentzer
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Khuzwayo C. Jere
- Malawi Liverpool Wellcome Programme, Blantyre, Malawi
- Institute of Infection, Veterinary, and Ecological Sciences, University of Liverpool, Liverpool, UK
- Kamuzu University of Health Sciences, Blantyre, Malawi
| |
Collapse
|
15
|
Samir A, Abdel-Moein KA, Zaher HM. Predominance of enterotoxigenic Escherichia coli among ESBL/plasmid-mediated AmpC-producing strains isolated from diarrheic foals: a public health concern. Acta Vet Scand 2024; 66:54. [PMID: 39363309 PMCID: PMC11448284 DOI: 10.1186/s13028-024-00774-6] [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: 01/20/2024] [Accepted: 09/11/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND The upsurge of diarrheagenic E. coli pathotypes carrying extended-spectrum beta-lactamases (ESBLs)/plasmid-mediated AmpC β-lactamase (pAmpC) among animals constitutes an emerging threat for humans and animals. This study investigated the burden of ESBL-/pAmpC-producing diarrheagenic E. coli among diarrheic foals and its potential public health implications. Rectal swabs were collected from 80 diarrheic foals. These swabs were processed to isolate and identify ESBL/pAmpC-producing E. coli using a selective culture medium, biochemical tests, phenotypic identification, and molecular identification of ESBL- and pAmpC-encoding genes. Moreover, all ESBL-/pAmpC-producing E. coli isolates were examined for different virulence genes related to diarrheagenic E. coli pathotypes. RESULTS Out of 80 examined foals, 26 (32.5%) were confirmed as ESBL-/pAmpC-producing E. coli, of which 14 (17.5%) animals carried only ESBL-producing E. coli, whereas 12 (15%) animals possessed ESBL-pAmpC-producing E. coli. The only detected diarrheagenic pathotype was enterotoxigenic, encoded by the heat-stable enterotoxin gene (ST) with a prevalence rate of 80.8% (21/26). The ST gene was further characterized where STa, STb, and STa + STb were found in one, four, and 16 strains, respectively. Moreover, all enterotoxigenic E. coli (ETEC) isolates exhibited a multidrug-resistance pattern. The phylogenetic analysis of 3 obtained partial STb sequences revealed high genetic relatedness to ETEC isolates retrieved from humans, conferring such sequences' public health significance. CONCLUSIONS These findings highlight that diarrheic foals could serve as a potential reservoir for multidrug-resistant ESBL-/pAmpC-producing enterotoxigenic E. coli.
Collapse
Affiliation(s)
- Ahmed Samir
- Department of Microbiology, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt
| | | | - Hala M Zaher
- Department of Zoonoses, Faculty of Veterinary Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
16
|
Berndsen ZT, Akhtar M, Thapa M, Vickers TJ, Schmitz A, Torres JL, Baboo S, Kumar P, Khatoon N, Sheikh A, Hamrick M, Diedrich JK, Martinez-Bartolome S, Garrett PT, Yates JR, Turner JS, Laird RM, Poly F, Porter CK, Copps J, Ellebedy AH, Ward AB, Fleckenstein JM. Repeat modules and N-linked glycans define structure and antigenicity of a critical enterotoxigenic E. coli adhesin. PLoS Pathog 2024; 20:e1012241. [PMID: 39283948 PMCID: PMC11463764 DOI: 10.1371/journal.ppat.1012241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 10/09/2024] [Accepted: 08/12/2024] [Indexed: 09/25/2024] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) cause hundreds of millions of cases of infectious diarrhea annually, predominantly in children from low-middle income regions. Notably, in children, as well as volunteers challenged with ETEC, diarrheal severity is significantly increased in blood group A (bgA) individuals. EtpA, is a secreted glycoprotein adhesin that functions as a blood group A lectin to promote critical interactions between ETEC and blood group A glycans on intestinal epithelia for effective bacterial adhesion and toxin delivery. EtpA is highly immunogenic resulting in robust antibody responses following natural infection and experimental challenge of volunteers with ETEC. To understand how EtpA directs ETEC-blood group A interactions and stimulates adaptive immunity, we mutated EtpA, mapped its glycosylation by mass-spectrometry (MS), isolated polyclonal (pAbs) and monoclonal antibodies (mAbs) from vaccinated mice and ETEC-infected volunteers, and determined structures of antibody-EtpA complexes by cryo-electron microscopy. Both bgA and mAbs that inhibited EtpA-bgA interactions and ETEC adhesion, bound to the C-terminal repeat domain highlighting this region as crucial for ETEC pathogen-host interaction. MS analysis uncovered extensive and heterogeneous N-linked glycosylation of EtpA and cryo-EM structures revealed that mAbs directly engage these unique glycan containing epitopes. Finally, electron microscopy-based polyclonal epitope mapping revealed antibodies targeting numerous distinct epitopes on N and C-terminal domains, suggesting that EtpA vaccination generates responses against neutralizing and decoy regions of the molecule. Collectively, we anticipate that these data will inform our general understanding of pathogen-host glycan interactions and adaptive immunity relevant to rational vaccine subunit design.
Collapse
Affiliation(s)
- Zachary T. Berndsen
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
| | - Marjahan Akhtar
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Mahima Thapa
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - Tim J. Vickers
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Aaron Schmitz
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - Jonathan L. Torres
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
| | - Sabyasachi Baboo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Pardeep Kumar
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Nazia Khatoon
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Alaullah Sheikh
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Melissa Hamrick
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
| | - Jolene K. Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Salvador Martinez-Bartolome
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Patrick T. Garrett
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - John R. Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | - Jackson S. Turner
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, United States of America
| | - Renee M. Laird
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
| | - Frédéric Poly
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
| | - Chad K. Porter
- Translational and Clinical Research Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
| | - Jeffrey Copps
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
| | - Ali H. Ellebedy
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine, Saint Louis, Missouri, United States of America
- Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, Missouri, United States of America
- The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, Missouri, United States of America
| | - Andrew B. Ward
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, California, United States of America
| | - James M. Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, United States of America
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, United States of America
- Medicine Service, Infectious Diseases, Veterans Affairs Health Care System, Saint Louis, Missouri, United States of America
| |
Collapse
|
17
|
Hasso-Agopsowicz M, Sparrow E, Cameron AM, Sati H, Srikantiah P, Gottlieb S, Bentsi-Enchill A, Le Doare K, Hamel M, Giersing BK, Hausdorff WP. The role of vaccines in reducing antimicrobial resistance: A review of potential impact of vaccines on AMR and insights across 16 vaccines and pathogens. Vaccine 2024; 42:S1-S8. [PMID: 38876836 DOI: 10.1016/j.vaccine.2024.06.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 05/01/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024]
Abstract
In 2019, an estimated 4.95 million deaths were linked to antimicrobial resistance (AMR). Vaccines can prevent many of these deaths by averting both drug-sensitive and resistant infections, reducing antibiotic usage, and lowering the likelihood of developing resistance genes. However, their role in mitigating AMR is currently underutilized. This article builds upon previous research that utilizes Vaccine Value Profiles-tools that assess the health, socioeconomic, and societal impact of pathogens-to inform vaccine development. We analyze the effects of 16 pathogens, covered by Vaccine Value Profiles, on AMR, and explore how vaccines could reduce AMR. The article also provides insights into vaccine development and usage. Vaccines are crucial in lessening the impact of infectious diseases and curbing the development of AMR. To fully realize their potential, vaccines must be more prominently featured in the overall strategy to combat AMR. This requires ongoing investment in research and development of new vaccines and the implementation of additional prevention and control measures to address this global threat effectively.
Collapse
Affiliation(s)
- Mateusz Hasso-Agopsowicz
- Vaccine Product & Delivery Research, Department of Immunization, Vaccines & Biologicals, World Health Organization, Geneva, Switzerland.
| | - Erin Sparrow
- Vaccine Product & Delivery Research, Department of Immunization, Vaccines & Biologicals, World Health Organization, Geneva, Switzerland
| | - Alexandra Meagan Cameron
- Global Coordination and Partnership (GCP), Antimicrobial Resistance Division, World Health Organization, Geneva, Switzerland
| | - Hatim Sati
- Global Coordination and Partnership (GCP), Antimicrobial Resistance Division, World Health Organization, Geneva, Switzerland
| | | | - Sami Gottlieb
- Department of Sexual and Reproductive Health and Research, World Health Organization, Geneva, Switzerland
| | - Adwoa Bentsi-Enchill
- Vaccine Product & Delivery Research, Department of Immunization, Vaccines & Biologicals, World Health Organization, Geneva, Switzerland
| | | | - Mary Hamel
- Vaccine Product & Delivery Research, Department of Immunization, Vaccines & Biologicals, World Health Organization, Geneva, Switzerland
| | - Birgitte K Giersing
- Vaccine Product & Delivery Research, Department of Immunization, Vaccines & Biologicals, World Health Organization, Geneva, Switzerland
| | - William P Hausdorff
- Center for Vaccine Access and Innovation, PATH, WA DC, USA; Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
18
|
Cassels FJ, Khalil I, Bourgeois AL, Walker RI. Special Issue on Enterotoxigenic Escherichia coli (ETEC) Vaccines: ETEC Infection and Vaccine-Mediated Immunity. Microorganisms 2024; 12:1087. [PMID: 38930469 PMCID: PMC11205504 DOI: 10.3390/microorganisms12061087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is the most prevalent bacterial pathogen causing young children to suffer acute watery diarrhea in Low- and Middle-Income Countries (LMICs) [...].
Collapse
Affiliation(s)
| | - Ibrahim Khalil
- Department of Global Health, University of Washington, Seattle, WA 98105, USA;
| | | | | |
Collapse
|
19
|
Berndsen ZT, Akhtar M, Thapa M, Vickers T, Schmitz A, Torres JL, Baboo S, Kumar P, Khatoom N, Sheikh A, Hamrick M, Diedrich JK, Martinez-Bartolome S, Garrett PT, Yates JR, Turner JS, Laird RM, Poly F, Porter CK, Copps J, Ellebedy AH, Ward AB, Fleckenstein JM. Repeat modules and N-linked glycans define structure and antigenicity of a critical enterotoxigenic E. coli adhesin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.08.593125. [PMID: 38766097 PMCID: PMC11100705 DOI: 10.1101/2024.05.08.593125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Enterotoxigenic Escherichia coli (ETEC) cause hundreds of millions of cases of infectious diarrhea annually, predominantly in children from low-middle income regions. Notably, in children, as well as human volunteers challenged with ETEC, diarrheal severity is significantly increased severity in blood group A (bgA) individuals. EtpA, is a secreted glycoprotein adhesin that functions as a blood group A lectin to promote critical interactions between ETEC and blood group A glycans on intestinal epithelia for effective bacterial adhesion and toxin delivery. EtpA is highly immunogenic resulting in robust antibody responses following natural infection and experimental challenge of human volunteers with ETEC. To understand how EtpA directs ETEC-blood group A interactions and stimulates adaptive immunity, we mutated EtpA, mapped its glycosylation by mass-spectrometry (MS), isolated polyclonal (pAbs) and monoclonal antibodies (mAbs) from vaccinated mice and ETEC-infected human volunteers, and determined structures of antibody-EtpA complexes by cryo-electron microscopy. Both bgA and mAbs that inhibited EtpA-bgA interactions and ETEC adhesion, bound to the C-terminal repeat domain highlighting this region as crucial for ETEC pathogen-host interaction. MS analysis uncovered extensive and heterogeneous N-linked glycosylation of EtpA and cryo-EM structures revealed that mAbs directly engage these unique glycan containing epitopes. Finally, electron microscopy-based polyclonal epitope mapping revealed antibodies targeting numerous distinct epitopes on N and C-terminal domains, suggesting that EtpA vaccination generates responses against neutralizing and decoy regions of the molecule. Collectively, we anticipate that these data will inform our general understanding of pathogen-host glycan interactions and adaptive immunity relevant to rational vaccine subunit design.
Collapse
Affiliation(s)
- Zachary T Berndsen
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Marjahan Akhtar
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Mahima Thapa
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA. Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Tim Vickers
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Aaron Schmitz
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA. Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Jonathan L Torres
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Sabyasachi Baboo
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Pardeep Kumar
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Nazia Khatoom
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Alaullah Sheikh
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Melissa Hamrick
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
| | - Jolene K Diedrich
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Patrick T Garrett
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - John R Yates
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Jackson S Turner
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA. Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Renee M Laird
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, USA
| | - Frédéric Poly
- Operationally Relevant Infections Department, Naval Medical Research Command (NMRC), Silver Spring, Maryland, USA
| | - Chad K Porter
- Naval Medical Research Command (NMRC), Silver Spring, Maryland, USA
- Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Jeffrey Copps
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - Ali H Ellebedy
- Department of Pathology and Immunology, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA. Center for Vaccines and Immunity to Microbial Pathogens, Washington University School of Medicine, St Louis, MO, USA and The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Andrew B Ward
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, USA
| | - James M Fleckenstein
- Department of Medicine, Division of Infectious Diseases, Washington University in Saint Louis, School of Medicine. Saint Louis, Missouri, USA
- Medicine Service, Infectious Diseases, Veterans Affairs Health Care System, Saint Louis Missouri, USA
| |
Collapse
|
20
|
von Mentzer A, Svennerholm AM. Colonization factors of human and animal-specific enterotoxigenic Escherichia coli (ETEC). Trends Microbiol 2024; 32:448-464. [PMID: 38052687 DOI: 10.1016/j.tim.2023.11.001] [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: 07/11/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 12/07/2023]
Abstract
Colonization factors (CFs) are major virulence factors of enterotoxigenic Escherichia coli (ETEC). This pathogen is among the most common causes of bacterial diarrhea in children in low- and middle-income countries, travelers, and livestock. CFs are major candidate antigens in vaccines under development as preventive measures against ETEC infections in humans and livestock. Recent molecular studies have indicated that newly identified CFs on human ETEC are closely related to animal ETEC CFs. Increased knowledge of pathogenic mechanisms, immunogenicity, regulation, and expression of ETEC CFs, as well as the possible spread of animal ETEC to humans, may facilitate the future development of ETEC vaccines for humans and animals. Here, we present an updated review of CFs in ETEC.
Collapse
Affiliation(s)
- Astrid von Mentzer
- Department of Microbiology and Immunology, Sahlgrenska Academy, University of Gothenburg, Sweden; Wellcome Sanger Institute, Hinxton, UK.
| | - Ann-Mari Svennerholm
- Department of Microbiology and Immunology, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
21
|
Sukwa N, Bosomprah S, Somwe P, Muyoyeta M, Mwape K, Chibesa K, Luchen CC, Silwamba S, Mulenga B, Munyinda M, Muzazu S, Chirwa M, Chibuye M, Simuyandi M, Chilengi R, Svennerholm AM. The Incidence and Risk Factors for Enterotoxigenic E. coli Diarrheal Disease in Children under Three Years Old in Lusaka, Zambia. Microorganisms 2024; 12:698. [PMID: 38674642 PMCID: PMC11051722 DOI: 10.3390/microorganisms12040698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/20/2024] [Accepted: 02/28/2024] [Indexed: 04/28/2024] Open
Abstract
This study aimed to estimate the incidence and risk factors for Enterotoxigenic Escherichia coli (ETEC) diarrhea. This was a prospective cohort study of children recruited in a household census. Children were enrolled if they were 36 months or below. A total of 6828 children were followed up passively for 12 months to detect episodes of ETEC diarrhea. Diarrheal stool samples were tested for ETEC using colony polymerase chain reaction (cPCR). Among the 6828 eligible children enrolled, a total of 1110 presented with at least one episode of diarrhea. The overall incidence of ETEC diarrhea was estimated as 2.47 (95% confidence interval (CI): 2.10-2.92) episodes per 100 child years. Children who were HIV-positive (adjusted Hazard ratio (aHR) = 2.14, 95% CI: 1.14 to 3.99; p = 0.017) and those whose source of drinking water was public tap/borehole/well (aHR = 2.45, 95% CI: 1.48 to 4.06; p < 0.002) were at increased risk of ETEC diarrhea. This study found that children whose mothers have at least senior secondary school education (aHR = 0.49, 95% CI: 0.29 to 0.83; p = 0.008) were at decreased risk of ETEC diarrhea. Our study emphasizes the need for integrated public health strategies focusing on water supply improvement, healthcare for persons living with HIV, and maternal education.
Collapse
Affiliation(s)
- Nsofwa Sukwa
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Samuel Bosomprah
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
- Department of Biostatistics, School of Public Health, University of Ghana, Accra P.O. Box LG13, Ghana
| | - Paul Somwe
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Monde Muyoyeta
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Kapambwe Mwape
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Kennedy Chibesa
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Charlie Chaluma Luchen
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Suwilanji Silwamba
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Bavin Mulenga
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Masiliso Munyinda
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Seke Muzazu
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Masuzyo Chirwa
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Mwelwa Chibuye
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Michelo Simuyandi
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Roma Chilengi
- Centre for Infectious Disease Research in Zambia (CIDRZ), Lusaka P.O. Box 34681, Zambia; (N.S.); (P.S.); (M.M.); (K.M.); (K.C.); (C.C.L.); (S.S.); (B.M.); (M.M.); (S.M.); (M.C.); (M.C.); (M.S.); (R.C.)
| | - Ann-Mari Svennerholm
- Department of Microbiology and Immunology, University of Gothenburg, 40530 Gothenburg, Sweden;
| |
Collapse
|
22
|
Pajuelo MJ, Noazin S, Cabrera L, Toledo A, Velagic M, Arias L, Ochoa M, Moulton LH, Saito M, Gilman RH, Chakraborty S. Epidemiology of enterotoxigenic Escherichia coli and impact on the growth of children in the first two years of life in Lima, Peru. Front Public Health 2024; 12:1332319. [PMID: 38584932 PMCID: PMC10995271 DOI: 10.3389/fpubh.2024.1332319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/11/2024] [Indexed: 04/09/2024] Open
Abstract
Background Enterotoxigenic E. coli (ETEC) is a leading cause of diarrheal morbidity and mortality in children, although the data on disease burden, epidemiology, and impact on health at the community level are limited. Methods In a longitudinal birth cohort study of 345 children followed until 24 months of age in Lima, Peru, we measured ETEC burden in diarrheal and non-diarrheal samples using quantitative PCR (LT, STh, and STp toxin genes), studied epidemiology and measured anthropometry in children. Results About 70% of children suffered from one or more ETEC diarrhea episodes. Overall, the ETEC incidence rate (IR) was 73 per 100 child-years. ETEC infections began early after birth causing 10% (8.9-11.1) ETEC-attributable diarrheal burden at the population level (PAF) in neonates and most of the infections (58%) were attributed to ST-ETEC [PAF 7.9% (1.9-13.5)] and LT + ST-ETEC (29%) of which all the episodes were associated with diarrhea. ETEC infections increased with age, peaking at 17% PAF (4.6-27.7%; p = 0.026) at 21 to 24 months. ST-ETEC was the most prevalent type (IR 32.1) with frequent serial infections in a child. The common colonization factors in ETEC diarrhea cases were CFA/I, CS12, CS21, CS3, and CS6, while in asymptomatic ETEC cases were CS12, CS6 and CS21. Only few (5.7%) children had repeated infections with the same combination of ETEC toxin(s) and CFs, suggested genotype-specific immunity from each infection. For an average ETEC diarrhea episode of 5 days, reductions of 0.060 weight-for-length z-score (0.007 to 0.114; p = 0.027) and 0.061 weight-for-age z-score (0.015 to 0.108; p = 0.009) were noted in the following 30 days. Conclusion This study showed that ETEC is a significant pathogen in Peruvian children who experience serial infections with multiple age-specific pathotypes, resulting in transitory growth impairment.
Collapse
Affiliation(s)
- Monica J. Pajuelo
- Laboratorio Microbiología Molecular – Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Sassan Noazin
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | | | - Angie Toledo
- Laboratorio Microbiología Molecular – Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Mirza Velagic
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Lucero Arias
- Laboratorio Microbiología Molecular – Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Mayra Ochoa
- Laboratorio Microbiología Molecular – Laboratorios de Investigación y Desarrollo, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Lawrence H. Moulton
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Mayuko Saito
- Department of Virology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Robert H. Gilman
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Subhra Chakraborty
- Department of International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
23
|
Pramudito TE, Desai K, Voigt C, Smid EJ, Schols HA. Dextran and levan exopolysaccharides from tempeh-associated lactic acid bacteria with bioactivity against enterotoxigenic Escherichia coli (ETEC). Carbohydr Polym 2024; 328:121700. [PMID: 38220337 DOI: 10.1016/j.carbpol.2023.121700] [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: 10/10/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/16/2024]
Abstract
Soybean tempeh contains bioactive carbohydrate that can reduce the severity of diarrhea by inhibiting enterotoxigenic Escherichia coli (ETEC) adhesion to mammalian epithelial cells. Lactic acid bacteria (LAB) are known to be present abundantly in soybean tempeh. Some LAB species can produce exopolysaccharides (EPS) with anti-adhesion bioactivity against ETEC but there has been no report of anti-adhesion bioactive EPS from tempeh-associated LAB. We isolated EPS-producing LAB from tempeh-related sources, identified them, unambiguously elucidated their EPS structure and assessed the bioactivity of their EPS against ETEC. Pediococcus pentosaceus TL, Leuconostoc mesenteroides WA and L. mesenteroides WN produced both dextran (α-1,6 linked glucan; >1000 kDa) and levan (β-2,6 linked fructan; 650-760 kDa) in varying amounts and Leuconostoc citreum TR produced gel-forming α-1,6-mixed linkage dextran (829 kDa). All four isolates produced EPS that could adhere to ETEC cells and inhibit auto-aggregation of ETEC. EPS-PpTL, EPS-LmWA and EPS-LmWN were more bioactive towards pig-associated ETEC K88 while EPS-LcTR was more bioactive against human-associated ETEC H10407. Our finding is the first to report on the bioactivity of dextran against ETEC. Tempeh is a promising source of LAB isolates that can produce bioactive EPS against ETEC adhesion and aggregation.
Collapse
Affiliation(s)
- Theodorus Eko Pramudito
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands; Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, Indonesia
| | - Krishna Desai
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands; Marie Curie Early Stage Researcher, NutriLeads B.V., the Netherlands
| | - Camiel Voigt
- Food Microbiology, Wageningen University & Research, the Netherlands
| | - Eddy J Smid
- Food Microbiology, Wageningen University & Research, the Netherlands
| | - Henk A Schols
- Laboratory of Food Chemistry, Wageningen University & Research, P.O. Box 17, 6700 AA Wageningen, the Netherlands.
| |
Collapse
|
24
|
Zhuang L, Gong J, Zhao Y, Yang J, Liu G, Zhao B, Song C, Zhang Y, Shen Q. Progress in methods for the detection of viable Escherichia coli. Analyst 2024; 149:1022-1049. [PMID: 38273740 DOI: 10.1039/d3an01750h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
Escherichia coli (E. coli) is a prevalent enteric bacterium and a necessary organism to monitor for food safety and environmental purposes. Developing efficient and specific methods is critical for detecting and monitoring viable E. coli due to its high prevalence. Conventional culture methods are often laborious and time-consuming, and they offer limited capability in detecting potentially harmful viable but non-culturable E. coli in the tested sample, which highlights the need for improved approaches. Hence, there is a growing demand for accurate and sensitive methods to determine the presence of viable E. coli. This paper scrutinizes various methods for detecting viable E. coli, including culture-based methods, molecular methods that target DNAs and RNAs, bacteriophage-based methods, biosensors, and other emerging technologies. The review serves as a guide for researchers seeking additional methodological options and aiding in the development of rapid and precise assays. Moving forward, it is anticipated that methods for detecting E. coli will become more stable and robust, ultimately contributing significantly to the improvement of food safety and public health.
Collapse
Affiliation(s)
- Linlin Zhuang
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong 212400, P. R. China.
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China.
| | - Jiansen Gong
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou 225125, P. R. China
| | - Ying Zhao
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China.
| | - Jianbo Yang
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong 212400, P. R. China.
| | - Guofang Liu
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong 212400, P. R. China.
| | - Bin Zhao
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong 212400, P. R. China.
| | - Chunlei Song
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong 212400, P. R. China.
| | - Yu Zhang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering & Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing 211102, P. R. China.
| | - Qiuping Shen
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong 212400, P. R. China.
| |
Collapse
|
25
|
Chakraborty S, Johura FT, Sultana M, Zhang X, Sadique A, George CM, Monira S, Sack DA, Sack RB, Alam M. Epidemiology of Enterotoxigenic Escherichia coli among Children and Adults Seeking Care at Hospitals in Two Geographically Distinct Rural Areas in Bangladesh. Microorganisms 2024; 12:359. [PMID: 38399763 PMCID: PMC10891752 DOI: 10.3390/microorganisms12020359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/01/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) infections undeniably continue to have substantial morbidity and mortality in younger children; however, limited data are available on the disease burden of older children and adults and on ETEC epidemiology by geographical location at the subnational level. Facility-based surveillance over the years was established to identify patients with ETEC diarrhea in two geographically distinct areas in rural Bangladesh, Chhatak in the north and Mathbaria in the southern coastal area. ETEC was highly prevalent in both areas, while the proportions, toxin types and colonization factors varied by location, season and age groups. Children < 5 years old and adults between 20 and 60 years old were at the highest risk of ETEC diarrhea which required urgent care. This study underscores the importance of capturing subnational and seasonal variations in ETEC epidemiology. ETEC vaccine developers and public health stakeholders may need to target adults between 20 and 60 years of age in addition to young children as new vaccines currently under development become licensed and introduction begins.
Collapse
Affiliation(s)
- Subhra Chakraborty
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; (F.-T.J.); (X.Z.); (C.M.G.); (D.A.S.); (R.B.S.)
| | - Fatema-Tuz Johura
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; (F.-T.J.); (X.Z.); (C.M.G.); (D.A.S.); (R.B.S.)
- International Centre for Diarrhoeal Disease Research, Dhaka 1212, Bangladesh; (M.S.); (A.S.); (S.M.); (M.A.)
| | - Marzia Sultana
- International Centre for Diarrhoeal Disease Research, Dhaka 1212, Bangladesh; (M.S.); (A.S.); (S.M.); (M.A.)
| | - Xueyan Zhang
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; (F.-T.J.); (X.Z.); (C.M.G.); (D.A.S.); (R.B.S.)
| | - Abdus Sadique
- International Centre for Diarrhoeal Disease Research, Dhaka 1212, Bangladesh; (M.S.); (A.S.); (S.M.); (M.A.)
| | - Christine M. George
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; (F.-T.J.); (X.Z.); (C.M.G.); (D.A.S.); (R.B.S.)
| | - Shirajum Monira
- International Centre for Diarrhoeal Disease Research, Dhaka 1212, Bangladesh; (M.S.); (A.S.); (S.M.); (M.A.)
| | - David A. Sack
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; (F.-T.J.); (X.Z.); (C.M.G.); (D.A.S.); (R.B.S.)
| | - Richard Bradley Sack
- Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; (F.-T.J.); (X.Z.); (C.M.G.); (D.A.S.); (R.B.S.)
| | - Munirul Alam
- International Centre for Diarrhoeal Disease Research, Dhaka 1212, Bangladesh; (M.S.); (A.S.); (S.M.); (M.A.)
| |
Collapse
|
26
|
Gutiérrez RL, Porter CK, Harro C, Talaat K, Riddle MS, DeNearing B, Brubaker J, Maciel M, Laird RM, Poole S, Chakraborty S, Maier N, Sack DA, Savarino SJ. Efficacy Evaluation of an Intradermally Delivered Enterotoxigenic Escherichia coli CF Antigen I Fimbrial Tip Adhesin Vaccine Coadministered with Heat-Labile Enterotoxin with LT(R192G) against Experimental Challenge with Enterotoxigenic E. coli H10407 in Healthy Adult Volunteers. Microorganisms 2024; 12:288. [PMID: 38399692 PMCID: PMC10892241 DOI: 10.3390/microorganisms12020288] [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: 12/12/2023] [Revised: 01/16/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Enterotoxigenic E. coli (ETEC) is a principal cause of diarrhea in travelers, deployed military personnel, and children living in low to middle-income countries. ETEC expresses a variety of virulence factors including colonization factors (CF) that facilitate adherence to the intestinal mucosa. We assessed the protective efficacy of a tip-localized subunit of CF antigen I (CFA/I), CfaE, delivered intradermally with the mutant E. coli heat-labile enterotoxin, LTR192G, in a controlled human infection model (CHIM). METHODS Three cohorts of healthy adult subjects were enrolled and given three doses of 25 μg CfaE + 100 ng LTR192G vaccine intradermally at 3-week intervals. Approximately 28 days after the last vaccination, vaccinated and unvaccinated subjects were admitted as inpatients and challenged with approximately 2 × 107 cfu of CFA/I+ ETEC strain H10407 following an overnight fast. Subjects were assessed for moderate-to-severe diarrhea for 5 days post-challenge. RESULTS A total of 52 volunteers received all three vaccinations; 41 vaccinated and 43 unvaccinated subjects were challenged and assessed for moderate-to-severe diarrhea. Naïve attack rates varied from 45.5% to 64.7% across the cohorts yielding an overall efficacy estimate of 27.8% (95% confidence intervals: -7.5-51.6%). In addition to reducing moderate-severe diarrhea rates, the vaccine significantly reduced loose stool output and overall ETEC disease severity. CONCLUSIONS This is the first study to demonstrate protection against ETEC challenge after intradermal vaccination with an ETEC adhesin. Further examination of the challenge methodology is necessary to address the variability in naïve attack rate observed among the three cohorts in the present study.
Collapse
Affiliation(s)
- Ramiro L. Gutiérrez
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
| | - Chad K. Porter
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
| | - Clayton Harro
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21224, USA (K.T.); (B.D.); (D.A.S.)
| | - Kawsar Talaat
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21224, USA (K.T.); (B.D.); (D.A.S.)
| | - Mark S. Riddle
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
| | - Barbara DeNearing
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21224, USA (K.T.); (B.D.); (D.A.S.)
| | - Jessica Brubaker
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21224, USA (K.T.); (B.D.); (D.A.S.)
| | - Milton Maciel
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Renee M. Laird
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Steven Poole
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD 20817, USA
| | - Subra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | | | - David A. Sack
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21224, USA (K.T.); (B.D.); (D.A.S.)
| | - Stephen J. Savarino
- Naval Medical Research Command, Silver Spring, MD 20910, USA; (R.L.G.); (M.S.R.); (R.M.L.); (S.P.); (S.J.S.)
| |
Collapse
|
27
|
Gerety MK, Kim DK, Carpenter RM, Ma JZ, Chisholm C, Taniuchi M, Islam MO, Pholwat S, Platts-Mills JA, Siraj MS, Billah SM, Haque R, Petri WA. Systemic inflammation, enteropathogenic E. Coli, and micronutrient insufficiencies in the first trimester as possible predictors of preterm birth in rural Bangladesh: a prospective study. BMC Pregnancy Childbirth 2024; 24:82. [PMID: 38267943 PMCID: PMC10807221 DOI: 10.1186/s12884-024-06266-9] [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: 09/13/2023] [Accepted: 01/10/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND An incomplete understanding of preterm birth is especially concerning for low-middle income countries, where preterm birth has poorer prognoses. While systemic proinflammatory processes are a reportedly normal component of gestation, excessive inflammation has been demonstrated as a risk factor for preterm birth. There is minimal research on the impact of excessive maternal inflammation in the first trimester on the risk of preterm birth in low-middle income countries specifically. METHODS Pregnant women were enrolled at the rural Bangladesh site of the National Institute of Child Health Global Network Maternal Newborn Health Registry. Serum samples were collected to measure concentrations of the inflammatory markers C-reactive protein (CRP) and Alpha-1-acid glycoprotein (AGP), and stool samples were collected and analyzed for enteropathogens. We examined associations of maternal markers in the first-trimester with preterm birth using logistic regression models. CRP and AGP were primarily modeled with a composite inflammation predictor. RESULTS Out of 376 singleton births analyzed, 12.5% were preterm. First trimester inflammation was observed in 58.8% of all births, and was significantly associated with increased odds of preterm birth (adjusted odds ratio [aOR] = 2.23; 95% confidence interval [CI]: 1.03, 5.16), independent of anemia. Maternal vitamin B12 insufficiency (aOR = 3.33; 95% CI: 1.29, 8.21) and maternal anemia (aOR = 2.56; 95% CI: 1.26, 5.17) were also associated with higher odds of preterm birth. Atypical enteropathogenic E. coli detection showed a significant association with elevated AGP levels and was significantly associated with preterm birth (odds ratio [OR] = 2.36; 95% CI: 1.21, 4.57), but not associated with CRP. CONCLUSIONS Inflammation, anemia, and vitamin B12 insufficiency in the first trimester were significantly associated with preterm birth in our cohort from rural Bangladesh. Inflammation and anemia were independent predictors of premature birth in this low-middle income setting where inflammation during gestation was widespread. Further research is needed to identify if infections such as enteropathogenic E. coli are a cause of inflammation in the first trimester, and if intervention for infection would decrease preterm birth.
Collapse
Affiliation(s)
- Meghan K Gerety
- Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Debora K Kim
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rebecca M Carpenter
- Department of Obstetrics and Gynecology, University of Texas Medical Branch, Galveston, TX, USA
| | - Jennie Z Ma
- Department of Public Health Sciences, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Christian Chisholm
- Department of Obstetrics and Gynecology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Mami Taniuchi
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Md Ohedul Islam
- The International Centre for Diarrhoeal Disease and Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Suporn Pholwat
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - James A Platts-Mills
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Md Shahjahan Siraj
- The International Centre for Diarrhoeal Disease and Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - Sk Masum Billah
- The International Centre for Diarrhoeal Disease and Research, Bangladesh (icddr,b), Dhaka, Bangladesh
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
| | - Rashidul Haque
- The International Centre for Diarrhoeal Disease and Research, Bangladesh (icddr,b), Dhaka, Bangladesh
| | - William A Petri
- Division of Infectious Diseases and International Health, University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
28
|
Kjellin J, Lee D, Steinsland H, Dwane R, Barth Vedoy O, Hanevik K, Koskiniemi S. Colicins and T6SS-based competition systems enhance enterotoxigenic E. coli (ETEC) competitiveness. Gut Microbes 2024; 16:2295891. [PMID: 38149626 PMCID: PMC10761095 DOI: 10.1080/19490976.2023.2295891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/13/2023] [Indexed: 12/28/2023] Open
Abstract
Diarrheal diseases are still a significant problem for humankind, causing approximately half a million deaths annually. To cause diarrhea, enteric bacterial pathogens must first colonize the gut, which is a niche occupied by the normal bacterial microbiota. Therefore, the ability of pathogenic bacteria to inhibit the growth of other bacteria can facilitate the colonization process. Although enterotoxigenic Escherichia coli (ETEC) is one of the major causative agents of diarrheal diseases, little is known about the competition systems found in and used by ETEC and how they contribute to the ability of ETEC to colonize a host. Here, we collected a set of 94 fully assembled ETEC genomes by performing whole-genome sequencing and mining the NCBI RefSeq database. Using this set, we performed a comprehensive search for delivered bacterial toxins and investigated how these toxins contribute to ETEC competitiveness in vitro. We found that type VI secretion systems (T6SS) were widespread among ETEC (n = 47). In addition, several closely related ETEC strains were found to encode Colicin Ia and T6SS (n = 8). These toxins provide ETEC competitive advantages during in vitro competition against other E. coli, suggesting that the role of T6SS as well as colicins in ETEC biology has until now been underappreciated.
Collapse
Affiliation(s)
- Jonas Kjellin
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Danna Lee
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Hans Steinsland
- CISMAC, Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Rachel Dwane
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Oda Barth Vedoy
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kurt Hanevik
- Department of Clinical Science, University of Bergen, Bergen, Norway
- National centre for Tropical Infectious Diseases, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Sanna Koskiniemi
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
29
|
Porter CK, Talaat KR, Isidean SD, Kardinaal A, Chakraborty S, Gutiérrez RL, Sack DA, Bourgeois AL. The Controlled Human Infection Model for Enterotoxigenic Escherichia coli. Curr Top Microbiol Immunol 2024; 445:189-228. [PMID: 34669040 DOI: 10.1007/82_2021_242] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The controlled human infection model (CHIM) for enterotoxigenic Escherichia coli (ETEC) has been instrumental in defining ETEC as a causative agent of acute watery diarrhea, providing insights into disease pathogenesis and resistance to illness, and enabling preliminary efficacy evaluations for numerous products including vaccines, immunoprophylactics, and drugs. Over a dozen strains have been evaluated to date, with a spectrum of clinical signs and symptoms that appear to replicate the clinical illness seen with naturally occurring ETEC. Recent advancements in the ETEC CHIM have enhanced the characterization of clinical, immunological, and microbiological outcomes. It is anticipated that omics-based technologies applied to ETEC CHIMs will continue to broaden our understanding of host-pathogen interactions and facilitate the development of primary and secondary prevention strategies.
Collapse
Affiliation(s)
- Chad K Porter
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA.
| | - Kawsar R Talaat
- Center for Immunization Research, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Sandra D Isidean
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA
- Henry M. Jackson Foundation, Bethesda, MD, 20817, USA
| | - Alwine Kardinaal
- NIZO Food Research, Ede, P.O. Box 20, 6710 BA EDE, Kernhemseweg 2, 6718 ZB EDE, The Netherlands
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Ramiro L Gutiérrez
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, 20910, USA
| | - David A Sack
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - A Louis Bourgeois
- PATH|Center for Vaccine Innovation and Access, 455 Massachusetts Avenue NW, Suite 1000, Washington, DC, 20001, USA
| |
Collapse
|
30
|
Hossain MJ, Svennerholm AM, Carlin N, D’Alessandro U, Wierzba TF. A Perspective on the Strategy for Advancing ETVAX ®, An Anti-ETEC Diarrheal Disease Vaccine, into a Field Efficacy Trial in Gambian Children: Rationale, Challenges, Lessons Learned, and Future Directions. Microorganisms 2023; 12:90. [PMID: 38257916 PMCID: PMC10819518 DOI: 10.3390/microorganisms12010090] [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: 11/15/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/24/2024] Open
Abstract
For the first time in over 20 years, an Enterotoxigenic Escherichia coli (ETEC) vaccine candidate, ETVAX®, has advanced into a phase 2b field efficacy trial for children 6-18 months of age in a low-income country. ETVAX® is an inactivated whole cell vaccine that has gone through a series of clinical trials to provide a rationale for the design elements of the Phase 2b trial. This trial is now underway in The Gambia and will be a precursor to an upcoming pivotal phase 3 trial. To reach this point, numerous findings were brought together to define factors such as safe and immunogenic doses for children, and the possible benefit of a mucosal adjuvant, double mutant labile toxin (dmLT). Considering the promising but still underexplored potential of inactivated whole cells in oral vaccination, we present a perspective compiling key observations from past ETVAX® trials that informed The Gambian trial design. This report will update the trial's status and explore future directions for ETEC vaccine trials. Our aim is to provide not only an update on the most advanced ETEC vaccine candidate but also to offer insights beneficial for the development of other much-needed oral whole-cell vaccines against enteric and other pathogens.
Collapse
Affiliation(s)
- M. Jahangir Hossain
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Banjul P.O. Box 273, The Gambia
| | - Ann-Mari Svennerholm
- Department of Microbiology and Immunology, Gothenburg University Research Institute (GUVAX), Gothenburg University, 40530 Gothenburg, Sweden
| | - Nils Carlin
- Scandinavian Biopharma, Industrivägen 1, 17148 Solna, Sweden
| | - Umberto D’Alessandro
- Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Banjul P.O. Box 273, The Gambia
| | - Thomas F. Wierzba
- Section on Infectious Diseases, Department of Internal Medicine, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| |
Collapse
|
31
|
Xu C, Fu F, She Y, Yang D, Peng K, Lin Y, Xu C. Development of a new candidate vaccine against piglet diarrhea caused by Escherichia coli. Open Life Sci 2023; 18:20220804. [PMID: 38196514 PMCID: PMC10775170 DOI: 10.1515/biol-2022-0804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/02/2023] [Accepted: 11/14/2023] [Indexed: 01/11/2024] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) is an important type of pathogenic bacteria that causes diarrhea in humans and young livestock. The pathogen has a high morbidity and mortality rate, resulting in significant economic losses in the pig industry. To effectively prevent piglet diarrhea, we developed a new tetravalent genetically engineered vaccine that specifically targets ETEC. To eliminate the natural toxin activity of ST1 enterotoxin and enhance the preventive effect of the vaccine, the mutated ST 1, K88ac, K99, and LT B genes were amplified by PCR and site-specific mutation techniques. The recombinant strain BL21(DE3)(pXKK3SL) was constructed and achieved high expression. Animal experiments showed that the inactivated vaccine had eliminated the natural toxin activity of ST1. The immune protection test demonstrated that the inclusion body and inactivated vaccine exhibited a positive immune effect. The protection rates of the inclusion body group and inactivated vaccine group were 96 and 98%, respectively, when challenged with 1 minimum lethal dose, indicating that the constructed K88ac-K99-3ST1-LTB vaccine achieved a strong immune effect. Additionally, the minimum immune doses for mice and pregnant sows were determined to be 0.2 and 2 mL, respectively. This study suggests that the novel K88ac-K99-3ST1-LTB vaccine has a wide immune spectrum and can prevent diarrhea caused by ETEC through enterotoxin and fimbrial pathways. The aforementioned research demonstrates that the K88ac-K99-3ST1-LTB vaccine offers a new genetically engineered vaccine that shows potential for preventing diarrhea in newborn piglets.
Collapse
Affiliation(s)
- Chongli Xu
- College of Medical Technology, Chongqing Medical and Pharmaceutical College, 82 Daxuecheng Road, Chongqing401331, PR China
| | - Fengyang Fu
- College of Medical Technology, Chongqing Medical and Pharmaceutical College, 82 Daxuecheng Road, Chongqing401331, PR China
| | - Yuhan She
- College of Medical Technology, Chongqing Medical and Pharmaceutical College, 82 Daxuecheng Road, Chongqing401331, PR China
| | - Danni Yang
- College of Medical Technology, Chongqing Medical and Pharmaceutical College, 82 Daxuecheng Road, Chongqing401331, PR China
| | - Kun Peng
- College of Medical Technology, Chongqing Medical and Pharmaceutical College, 82 Daxuecheng Road, Chongqing401331, PR China
| | - Yimin Lin
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing400030, PR China
| | - ChongBo Xu
- School of Biology and Agriculture, Shaoguan University, Shaoguan512005, PR China
| |
Collapse
|
32
|
Lu X, Zhang M, Ma Y, Li G, Zhao X, Qian W. Protective effect of Limosilactobacillus reuteri-fermented yogurt on mouse intestinal barrier injury induced by enterotoxigenic Escherichia coli. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:7494-7505. [PMID: 37411001 DOI: 10.1002/jsfa.12836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/25/2023] [Accepted: 07/07/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND Enterotoxigenic Escherichia coli (ETEC) is a pathogen that causes traveler's diarrhea, for which an effective vaccine is lacking. Previous studies showed that Limosilactobacillus reuteri could inhibit E. coli, effectively increase the expression of its tight junction protein, and reduce the adhesion of ETEC to the intestinal epithelial Caco-2 cell line. In this study, three kinds of yogurt with different starter cultures were first prepared: Lm. reuteri yogurt (fermented by Lm. reuteri alone), traditional yogurt (fermented by Streptococcus thermophilus and Lactobacillus delbrueckii subsp. bulgaricus at a ratio of 1:1) and mixed yogurt (fermented by Lm. reuteri, S. thermophilus and L. delbrueckii subsp. bulgaricus at a ratio of 1:1:1). The physiological properties, oxidative stress, intestinal barrier function, tight junction protein, pathological conditions and intestinal microbiota composition were investigated. RESULTS The data showed that Lm. reuteri-fermented yogurt pregavage could effectively alleviate the intestinal barrier impairment caused by ETEC in mice. It alleviated intestinal villus shortening and inflammatory cell infiltration, decreased plasma diamine oxidase concentration and increased claudin-1 and occludin expression in the jejunum of ETEC-infected mice. In addition, Lm. reuteri-fermented yogurt significantly reduced the ETEC load in fecal samples, reversed the increase in Pseudomonadota abundance and decreased Bacteroidota abundance caused by ETEC infection. Furthermore, the composition of the intestinal microbiota could maintain a stable state similar to that in healthy mice. CONCLUSION These findings indicate that Lm. reuteri-fermented yogurt could alleviate intestinal barrier damage, inhibit ETEC growth and maintain the stability of the intestinal microbiota during ETEC infection. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Xi Lu
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Mingxin Zhang
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Yuzhe Ma
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Guohua Li
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Xin Zhao
- School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an, China
| | - Weisheng Qian
- Tangdu Hospital, Air Force Military Medical University, Xi'an, China
| |
Collapse
|
33
|
Héma A, Sermé SS, Sawadogo J, Diarra A, Barry A, Ouédraogo AZ, Nébié I, Tiono AB, Houard S, Chakraborty S, Ouédraogo A, Sirima SB. Contribution of the Rapid LAMP-Based Diagnostic Test (RLDT) to the Evaluation of Enterotoxigenic Escherichia coli (ETEC) and Shigella in Childhood Diarrhea in the Peri-Urban Area of Ouagadougou, Burkina Faso. Microorganisms 2023; 11:2809. [PMID: 38004821 PMCID: PMC10673293 DOI: 10.3390/microorganisms11112809] [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: 09/23/2023] [Revised: 11/12/2023] [Accepted: 11/17/2023] [Indexed: 11/26/2023] Open
Abstract
The estimates of enterotoxigenic Escherichia coli (ETEC) and Shigella burden in developing countries are limited by the lack of rapid, accessible, and sensitive diagnostics and surveillance tools. We used a "Rapid LAMP based Diagnostic Test (RLDT)" to detect ETEC and Shigella in diarrheal and non-diarrheal stool samples from a 12-month longitudinal cohort of children under five years of age in a peri-urban area of Ouagadougou in Burkina Faso (West Africa). To allow comparison with the RLDT-Shigella results, conventional culture methods were used to identify Shigella strains in the stool samples. As conventional culture alone cannot detect ETEC cases, a subset of E. coli-like colonies was tested using conventional PCR to detect ETEC toxins genes. Of the 165 stool samples analyzed for ETEC, 24.9% were positive when using RLDT against 4.2% when using culture followed by PCR. ETEC toxin distribution when using RLDT was STp 17.6% (29/165), LT 11.5% (19/165), and STh 8.5% (14/165). Of the 263 specimens tested for Shigella, 44.8% were positive when using RLDT against 23.2% when using culture. The sensitivity and specificity of the RLDT compared to culture (followed by PCR for ETEC) were 93.44% and 69.8% for Shigella and 83.7% and 77.9% for ETEC, respectively. This study indicates that both Shigella and ETEC are substantially underdiagnosed when using conventional culture and highlights the potential contribution of the new RLDT method to improve enteric disease burden estimation and to guide future efforts to prevent and control bacterial enteric infection and disease.
Collapse
Affiliation(s)
- Alimatou Héma
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 06 BP 10248, Burkina Faso; (S.S.S.); (J.S.); (A.D.); (A.B.); (A.Z.O.); (I.N.); (A.B.T.); (A.O.); (S.B.S.)
| | - Samuel S. Sermé
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 06 BP 10248, Burkina Faso; (S.S.S.); (J.S.); (A.D.); (A.B.); (A.Z.O.); (I.N.); (A.B.T.); (A.O.); (S.B.S.)
| | - Jean Sawadogo
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 06 BP 10248, Burkina Faso; (S.S.S.); (J.S.); (A.D.); (A.B.); (A.Z.O.); (I.N.); (A.B.T.); (A.O.); (S.B.S.)
| | - Amidou Diarra
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 06 BP 10248, Burkina Faso; (S.S.S.); (J.S.); (A.D.); (A.B.); (A.Z.O.); (I.N.); (A.B.T.); (A.O.); (S.B.S.)
| | - Aissata Barry
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 06 BP 10248, Burkina Faso; (S.S.S.); (J.S.); (A.D.); (A.B.); (A.Z.O.); (I.N.); (A.B.T.); (A.O.); (S.B.S.)
| | - Amidou Z. Ouédraogo
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 06 BP 10248, Burkina Faso; (S.S.S.); (J.S.); (A.D.); (A.B.); (A.Z.O.); (I.N.); (A.B.T.); (A.O.); (S.B.S.)
| | - Issa Nébié
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 06 BP 10248, Burkina Faso; (S.S.S.); (J.S.); (A.D.); (A.B.); (A.Z.O.); (I.N.); (A.B.T.); (A.O.); (S.B.S.)
| | - Alfred B. Tiono
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 06 BP 10248, Burkina Faso; (S.S.S.); (J.S.); (A.D.); (A.B.); (A.Z.O.); (I.N.); (A.B.T.); (A.O.); (S.B.S.)
| | - Sophie Houard
- European Vaccine Initiative (EVI), Universitäts Klinikum Heidelberg Vossstrasse 2, Geb. 4040, 69115 Heidelberg, Germany;
| | - Subhra Chakraborty
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA;
| | - Alphonse Ouédraogo
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 06 BP 10248, Burkina Faso; (S.S.S.); (J.S.); (A.D.); (A.B.); (A.Z.O.); (I.N.); (A.B.T.); (A.O.); (S.B.S.)
| | - Sodiomon B. Sirima
- Groupe de Recherche Action en Santé (GRAS), Ouagadougou 06 BP 10248, Burkina Faso; (S.S.S.); (J.S.); (A.D.); (A.B.); (A.Z.O.); (I.N.); (A.B.T.); (A.O.); (S.B.S.)
| |
Collapse
|
34
|
Kantele A, Riekkinen M, Jokiranta TS, Pakkanen SH, Pietilä JP, Patjas A, Eriksson M, Khawaja T, Klemets P, Marttinen K, Siikamäki H, Lundgren A, Holmgren J, Lissmats A, Carlin N, Svennerholm AM. Safety and immunogenicity of ETVAX®, an oral inactivated vaccine against enterotoxigenic Escherichia coli diarrhoea: a double-blinded, randomized, placebo-controlled trial amongst Finnish travellers to Benin, West Africa. J Travel Med 2023; 30:taad045. [PMID: 37099803 PMCID: PMC10658657 DOI: 10.1093/jtm/taad045] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/31/2023] [Accepted: 03/28/2023] [Indexed: 04/28/2023]
Abstract
BACKGROUND No licensed human vaccines are available against enterotoxigenic Escherichia coli (ETEC), a major diarrhoeal pathogen affecting children in low- and middle-income countries and foreign travellers alike. ETVAX®, a multivalent oral whole-cell vaccine containing four inactivated ETEC strains and the heat-labile enterotoxin B subunit (LTB), has proved promising in Phase 1 and Phase 1/ 2 studies. METHODS We conducted a Phase 2b double-blinded, randomized, placebo-controlled trial amongst Finnish travellers to Benin, West Africa. This report presents study design and safety and immunogenicity data. Volunteers aged 18-65 years were randomized 1:1 to receive ETVAX® or placebo. They visited Benin for 12 days, provided stool and blood samples and completed adverse event (AE) forms. IgA and IgG antibodies to LTB and O78 lipopolysaccharide (LPS) were measured by electrochemiluminescence. RESULTS The AEs did not differ significantly between vaccine (n = 374) and placebo (n = 375) recipients. Of the solicited AEs, loose stools/diarrhoea (26.7/25.9%) and stomach ache (23.0/20.0%) were reported most commonly. Of all possibly/probably vaccine-related AEs, the most frequent were gastrointestinal symptoms (54.0/48.8%) and nervous system disorders (20.3/25.1%). Serious AEs were recorded for 4.3/5.6%, all unlikely to be vaccine related. Amongst the ETVAX® recipients, LTB-specific IgA antibodies increased 22-fold. For the 370/372 vaccine/placebo recipients, the frequency of ≥2-fold increases against LTB was 81/2.4%, and against O78 LPS 69/2.7%. The majority of ETVAX® recipients (93%) responded to either LTB or O78. CONCLUSIONS This Phase 2b trial is the largest on ETVAX® undertaken amongst travellers to date. ETVAX® showed an excellent safety profile and proved strongly immunogenic, which encourages the further development of this vaccine.
Collapse
Affiliation(s)
- Anu Kantele
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Unit, University of Helsinki, Helsinki, Finland
- Travel Clinic, Aava Medical Center, Helsinki, Finland
| | - Marianna Riekkinen
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Unit, University of Helsinki, Helsinki, Finland
- Travel Clinic, Aava Medical Center, Helsinki, Finland
| | - T Sakari Jokiranta
- United Medix Laboratories/Synlab Finland Ltd, Helsinki, Finland
- Medicum, Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
- Mobidiag Ltd, Espoo, Finland
| | - Sari H Pakkanen
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Unit, University of Helsinki, Helsinki, Finland
| | - Jukka-Pekka Pietilä
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Unit, University of Helsinki, Helsinki, Finland
- Travel Clinic, Aava Medical Center, Helsinki, Finland
| | - Anu Patjas
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Unit, University of Helsinki, Helsinki, Finland
- Travel Clinic, Aava Medical Center, Helsinki, Finland
| | - Mari Eriksson
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
| | - Tamim Khawaja
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Unit, University of Helsinki, Helsinki, Finland
| | - Peter Klemets
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
| | - Kati Marttinen
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
| | - Heli Siikamäki
- Meilahti Vaccine Research Center, MeVac, University of Helsinki and Department of Infectious Diseases, Inflammation Center, HUS, Helsinki University Hospital, Helsinki, Finland
| | - Anna Lundgren
- Gothenburg University Vaccine Research Institute, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Jan Holmgren
- Gothenburg University Vaccine Research Institute, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Ann-Mari Svennerholm
- Gothenburg University Vaccine Research Institute, Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
35
|
Khalil I, Anderson JD, Bagamian KH, Baqar S, Giersing B, Hausdorff WP, Marshall C, Porter CK, Walker RI, Bourgeois AL. Vaccine value profile for enterotoxigenic Escherichia coli (ETEC). Vaccine 2023; 41 Suppl 2:S95-S113. [PMID: 37951695 DOI: 10.1016/j.vaccine.2023.02.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 11/28/2022] [Accepted: 02/05/2023] [Indexed: 11/14/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is one of the leading bacterial causes of diarrhoea, especially among children in low-resource settings, and travellers and military personnel from high-income countries. WHO's primary strategic goal for ETEC vaccine development is to develop a safe, effective, and affordable ETEC vaccine that reduces mortality and morbidity due to moderate-to-severe diarrhoeal disease in infants and children under 5 years of age in LMICs, as well as the long-term negative health impact on infant physical and cognitive development resulting from infection with this enteric pathogen. An effective ETEC vaccine will also likely reduce the need for antibiotic treatment and help limit the further emergence of antimicrobial resistance bacterial pathogens. The lead ETEC vaccine candidate, ETVAX, has shown field efficacy in travellers and has moved into field efficacy testing in LMIC infants and children. A Phase 3 efficacy study in LMIC infants is projected to start in 2024 and plans for a Phase 3 trial in travellers are under discussion with the U.S. FDA. Licensing for both travel and LMIC indications is projected to be feasible in the next 5-8 years. Given increasing recognition of its negative impact on child health and development in LMICs and predominance as the leading etiology of travellers' diarrhoea (TD), a standalone vaccine for ETEC is more cost-effective than vaccines targeting other TD pathogens, and a viable commercial market also exists. In contrast, combination of an ETEC vaccine with other vaccines for childhood pathogens in LMICs would maximize protection in a more cost-effective manner than a series of stand-alone vaccines. This 'Vaccine Value Profile' (VVP) for ETEC is intended to provide a high-level, holistic assessment of available data to inform the potential public health, economic and societal value of pipeline vaccines and vaccine-like products. This VVP was developed by a working group of subject matter experts from academia, non-profit organizations, public private partnerships, and multi-lateral organizations. All contributors have extensive expertise on various elements of the ETEC VVP and collectively aimed to identify current research and knowledge gaps. The VVP was developed using only existing and publicly available information.
Collapse
Affiliation(s)
- Ibrahim Khalil
- Department of Global Health, University of Washington, Seattle, WA 98195, USA.
| | - John D Anderson
- Bagamian Scientific Consulting, LLC, Gainesville, FL 32601, USA; Office of Health Affairs, West Virginia University, Morgantown, WV 26505, USA
| | - Karoun H Bagamian
- Bagamian Scientific Consulting, LLC, Gainesville, FL 32601, USA; Department of Environmental and Global Health, University of Florida, Gainesville, FL 32603, USA
| | - Shahida Baqar
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Birgitte Giersing
- Department of Immunization, Vaccines and Biologicals (IVB), World Health Organization (WHO), Geneva, Switzerland
| | - William P Hausdorff
- PATH, Center for Vaccine Innovation and Access, 455 Massachusetts Ave NW, Washington, DC 20001 USA; Faculty of Medicine, Université Libre de Bruxelles, Brussels 1070, Belgium
| | - Caroline Marshall
- Department of Immunization, Vaccines and Biologicals (IVB), World Health Organization (WHO), Geneva, Switzerland
| | - Chad K Porter
- Directorate for DoD Infectious Diseases Research, Naval Medical Research Command, Silver Spring, MD 20190, USA
| | - Richard I Walker
- PATH, Center for Vaccine Innovation and Access, 455 Massachusetts Ave NW, Washington, DC 20001 USA
| | - A Louis Bourgeois
- PATH, Center for Vaccine Innovation and Access, 455 Massachusetts Ave NW, Washington, DC 20001 USA
| |
Collapse
|
36
|
Sukwa N, Mubanga C, Hatyoka LM, Chilyabanyama ON, Chibuye M, Mundia S, Munyinda M, Kamuti E, Siyambango M, Badiozzaman S, Bosomprah S, Carlin N, Kaim J, Sjöstrand B, Simuyandi M, Chilengi R, Svennerholm AM. Safety, tolerability, and immunogenicity of an oral inactivated ETEC vaccine (ETVAX®) with dmLT adjuvant in healthy adults and children in Zambia: An age descending randomised, placebo-controlled trial. Vaccine 2023; 41:6884-6894. [PMID: 37838479 DOI: 10.1016/j.vaccine.2023.09.052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 08/07/2023] [Accepted: 09/23/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND Enterotoxigenic Escherichia coli (ETEC) is an important cause of moderate to severe diarrhoea in children for which there is no licensed vaccine. We evaluated ETVAX®, an oral, inactivated ETEC vaccine containing four E. coli strains over-expressing the major colonization factors CFA/I, CS3, CS5, and CS6, a toxoid (LCTBA) and double mutant heat-labile enterotoxin (dmLT) adjuvant for safety, tolerability, and immunogenicity. METHODS A double-blind, placebo-controlled, age-descending, dose-finding trial was undertaken in 40 adults, 60 children aged 10-23 months, and 146 aged 6-9 months. Adults received one full dose of ETVAX® and children received 3 doses of either 1/4 or 1/8 dose. Safety was evaluated as solicited and unsolicited events for 7 days following vaccination. Immunogenicity was assessed by evaluation of plasma IgA antibody responses to CFA/I, CS3, CS5, CS6, and LTB, and IgG responses to LTB. RESULTS Solicited adverse events were mostly mild or moderate with only 2 severe fever reports which were unrelated to the vaccine. The most common events were abdominal pain in adults (26.7 % in vaccinees vs 20 % in placebos), and fever in children aged 6-9 months (44 % vs 54 %). Dosage, number of vaccinations and decreasing age had no influence on severity or frequency of adverse events. The vaccine induced plasma IgA and IgG responses against LTB in 100 % of the adults and 80-90 % of the children. In the 6-23 months cohort, IgA responses to more than 3 vaccine antigens after 3 doses determined as ≥2-fold rise was significantly higher for 1/4 dose compared to placebo (56.7 % vs 27.2 %, p = 0.01). In the 6-9 months cohort, responses to the 1/4 dose were significantly higher than 1/8 dose after 3 rather than 2 doses. CONCLUSION ETVAX® was safe, tolerable, and immunogenic in Zambian adults and children. The 1/4 dose induced significantly stronger IgA responses and is recommended for evaluation of protection in children. CLINICAL TRIALS REGISTRATION The trial is registered with the Pan African Clinical Trials Registry (PACTR Ref. 201905764389804) and a description of this clinical trial is available on: https://pactr.samrc.ac.za/Trial Design.
Collapse
Affiliation(s)
- Nsofwa Sukwa
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka, Zambia.
| | - Cynthia Mubanga
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka, Zambia; Division of Medical Microbiology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, South Africa
| | - Luiza M Hatyoka
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| | - Obvious N Chilyabanyama
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| | - Mwelwa Chibuye
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| | - Samson Mundia
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| | - Masiliso Munyinda
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| | - Ethel Kamuti
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| | - Muyunda Siyambango
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| | - Sharif Badiozzaman
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| | - Samuel Bosomprah
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka, Zambia; Department of Biostatistics, School of Public Health, University of Ghana, Accra, Ghana
| | | | - Joanna Kaim
- Department of Microbiology and Immunology, University of Gothenburg, Sweden
| | | | - Michelo Simuyandi
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| | - Roma Chilengi
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka, Zambia
| | | |
Collapse
|
37
|
Piroozkhah M, Aghajani A, Jalali P, Shahmoradi A, Piroozkhah M, Tadlili Y, Salehi Z. Guanylate cyclase-C Signaling Axis as a theragnostic target in colorectal cancer: a systematic review of literature. Front Oncol 2023; 13:1277265. [PMID: 37927469 PMCID: PMC10623427 DOI: 10.3389/fonc.2023.1277265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/28/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction Colorectal cancer (CRC) is a devastating disease that affects millions of people worldwide. Recent research has highlighted the crucial role of the guanylate cyclase-C (GC-C) signaling axis in CRC, from the early stages of tumorigenesis to disease progression. GC-C is activated by endogenous peptides guanylin (GU) and uroguanylin (UG), which are critical in maintaining intestinal fluid homeostasis. However, it has been found that these peptides may also contribute to the development of CRC. This systematic review focuses on the latest research on the GC-C signaling axis in CRC. Methods According to the aim of the study, a systematic literature search was conducted on Medline and PubMed databases. Ultimately, a total of 40 articles were gathered for the systematic review. Results Our systematic literature search revealed that alterations in GC-C signaling compartments in CRC tissue have demonstrated potential as diagnostic, prognostic, and therapeutic markers. This research highlights a potential treatment for CRC by targeting the GC-C signaling axis. Promising results from recent studies have explored the use of this signaling axis to develop new vaccines and chimeric antigen receptors that may be used in future clinical trials. Conclusion The findings presented in this review provide compelling evidence that targeting the GC-C signaling axis may be an advantageous approach for treating CRC.
Collapse
Affiliation(s)
- Moein Piroozkhah
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Aghajani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arvin Shahmoradi
- Department of Laboratory Medicine, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mobin Piroozkhah
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Younes Tadlili
- Department of Molecular Cell Biology, Microbiology Trend, Faculty of Basic Sciences, Islamic Azad University, Central Tehran Branch, Tehran, Iran
| | - Zahra Salehi
- Hematology-Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
38
|
Kim CL, Agampodi S, Marks F, Kim JH, Excler JL. Mitigating the effects of climate change on human health with vaccines and vaccinations. Front Public Health 2023; 11:1252910. [PMID: 37900033 PMCID: PMC10602790 DOI: 10.3389/fpubh.2023.1252910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/04/2023] [Indexed: 10/31/2023] Open
Abstract
Climate change represents an unprecedented threat to humanity and will be the ultimate challenge of the 21st century. As a public health consequence, the World Health Organization estimates an additional 250,000 deaths annually by 2030, with resource-poor countries being predominantly affected. Although climate change's direct and indirect consequences on human health are manifold and far from fully explored, a growing body of evidence demonstrates its potential to exacerbate the frequency and spread of transmissible infectious diseases. Effective, high-impact mitigation measures are critical in combating this global crisis. While vaccines and vaccination are among the most cost-effective public health interventions, they have yet to be established as a major strategy in climate change-related health effect mitigation. In this narrative review, we synthesize the available evidence on the effect of climate change on vaccine-preventable diseases. This review examines the direct effect of climate change on water-related diseases such as cholera and other enteropathogens, helminthic infections and leptospirosis. It also explores the effects of rising temperatures on vector-borne diseases like dengue, chikungunya, and malaria, as well as the impact of temperature and humidity on airborne diseases like influenza and respiratory syncytial virus infection. Recent advances in global vaccine development facilitate the use of vaccines and vaccination as a mitigation strategy in the agenda against climate change consequences. A focused evaluation of vaccine research and development, funding, and distribution related to climate change is required.
Collapse
Affiliation(s)
- Cara Lynn Kim
- International Vaccine Institute, Seoul, Republic of Korea
| | - Suneth Agampodi
- International Vaccine Institute, Seoul, Republic of Korea
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Florian Marks
- International Vaccine Institute, Seoul, Republic of Korea
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Madagascar Institute for Vaccine Research, University of Antananarivo, Antananarivo, Madagascar
- Heidelberg Institute of Global Health, University of Heidelberg, Heidelberg, Germany
| | - Jerome H. Kim
- International Vaccine Institute, Seoul, Republic of Korea
- College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | | |
Collapse
|
39
|
Svennerholm AM, Lundgren A. Developments in oral enterotoxigenic Escherichia coli vaccines. Curr Opin Immunol 2023; 84:102372. [PMID: 37523966 DOI: 10.1016/j.coi.2023.102372] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/11/2023] [Accepted: 07/04/2023] [Indexed: 08/02/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a leading cause of diarrhea in children in developing countries and in travelers. WHO has affirmed ETEC as a priority vaccine target, but there is no licensed ETEC vaccine available yet. We here describe recent, promising developments of different live, inactivated, and subunit ETEC candidate vaccines expressing or containing nontoxic enterotoxin and/or colonization factor antigens with a focus on oral vaccines. Many of the ETEC candidate vaccines have been tested in clinical trials for safety and immunogenicity and some of them also for protective efficacy in field trials or in challenge studies.
Collapse
Affiliation(s)
- Ann-Mari Svennerholm
- Dept. of Microbiology and Immunology, Inst. of Biomedicine, University of Gothenburg, Sweden.
| | - Anna Lundgren
- Dept. of Microbiology and Immunology, Inst. of Biomedicine, University of Gothenburg, Sweden
| |
Collapse
|
40
|
Maier N, Grahek SL, Halpern J, Restrepo S, Troncoso F, Shimko J, Torres O, Belkind-Gerson J, Sack DA, Svennerholm AM, Gustafsson B, Sjöstrand B, Carlin N, Bourgeois AL, Porter CK. Efficacy of an Enterotoxigenic Escherichia coli (ETEC) Vaccine on the Incidence and Severity of Traveler's Diarrhea (TD): Evaluation of Alternative Endpoints and a TD Severity Score. Microorganisms 2023; 11:2414. [PMID: 37894071 PMCID: PMC10609384 DOI: 10.3390/microorganisms11102414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
The efficacy of an Oral Whole Cell ETEC Vaccine (OEV) against Travelers' Diarrhea (TD) was reexamined using novel outcome and immunologic measures. More specifically, a recently developed disease severity score and alternative clinical endpoints were evaluated as part of an initial validation effort to access the efficacy of a vaccine intervention for the first time in travelers to an ETEC endemic area. A randomized, double-blind, placebo-controlled trial followed travelers to Guatemala or Mexico up to 28 days after arrival in the country following vaccination (two doses two weeks apart) with an ETEC vaccine. Fecal samples were collected upon arrival, departure, and during TD for pathogen identification. Serum was collected in a subset of subjects to determine IgA cholera toxin B subunit (CTB) antibody titers upon their arrival in the country. The ETEC vaccine's efficacy, utilizing a TD severity score and other alternative endpoints, including the relationship between antibody levels and TD risk, was assessed and compared to the per-protocol primary efficacy endpoint. A total of 1435 subjects completed 7-28 days of follow-up and had available data. Vaccine efficacy was higher against more severe (≥5 unformed stools/24 h) ETEC-attributable TD and when accounting for immunologic take (PE ≥ 50%; p < 0.05). The vaccine protected against less severe (3 and 4 unformed stools/24 h) ETEC-attributable TD when accounting for symptom severity or change in activity (PE = 76.3%, p = 0.01). Immunologic take of the vaccine was associated with a reduced risk of infection with ETEC and other enteric pathogens, and with lower TD severity. Clear efficacy was observed among vaccinees with a TD score of ≥4 or ≥5, regardless of immunologic take (PE = 72.0% and 79.0%, respectively, p ≤ 0.03). The vaccine reduced the incidence and severity of ETEC, and this warrants accelerated evaluation of the improved formulation (designated ETVAX), currently undergoing advanced field testing. Subjects with serum IgA titers to CTB had a lower risk of infection with ETEC and Campylobacter jejuni/coli. Furthermore, the TD severity score provided a more robust descriptor of disease severity and should be included as an endpoint in future studies.
Collapse
Affiliation(s)
| | - Shannon L. Grahek
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.L.G.); (J.H.); (S.R.); (F.T.); (J.S.); (D.A.S.)
| | - Jane Halpern
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.L.G.); (J.H.); (S.R.); (F.T.); (J.S.); (D.A.S.)
| | - Suzanne Restrepo
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.L.G.); (J.H.); (S.R.); (F.T.); (J.S.); (D.A.S.)
| | - Felipe Troncoso
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.L.G.); (J.H.); (S.R.); (F.T.); (J.S.); (D.A.S.)
| | - Janet Shimko
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.L.G.); (J.H.); (S.R.); (F.T.); (J.S.); (D.A.S.)
| | - Olga Torres
- Laboratorio Diagnostico Molecular, Guatemala City 01009, Guatemala;
| | | | - David A. Sack
- Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (S.L.G.); (J.H.); (S.R.); (F.T.); (J.S.); (D.A.S.)
| | - Ann-Mari Svennerholm
- Department of Microbiology and Immunology, University of Gothenburg, 405 30 Gothenburg, Sweden;
| | - Björn Gustafsson
- Scandinavian Biopharma Holding AB, 171 48 Stockholm, Sweden (B.S.); (N.C.)
| | - Björn Sjöstrand
- Scandinavian Biopharma Holding AB, 171 48 Stockholm, Sweden (B.S.); (N.C.)
| | - Nils Carlin
- Scandinavian Biopharma Holding AB, 171 48 Stockholm, Sweden (B.S.); (N.C.)
| | | | - Chad K. Porter
- Naval Medical Research Command, Silver Spring, MD 20910, USA;
| |
Collapse
|
41
|
Kuhlmann FM, Grigura V, Vickers TJ, Prouty MG, Iannotti LL, Dulience SJL, Fleckenstein JM. Seroprevalence Study of Conserved Enterotoxigenic Escherichia coli Antigens in Globally Diverse Populations. Microorganisms 2023; 11:2221. [PMID: 37764065 PMCID: PMC10536235 DOI: 10.3390/microorganisms11092221] [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: 07/18/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Enterotoxigenic Escherichia coli (ETEC) are common causes of infectious diarrhea among young children of low-and middle-income countries (LMICs) and travelers to these regions. Despite their significant contributions to the morbidity and mortality associated with childhood and traveler's diarrhea, no licensed vaccines are available. Current vaccine strategies may benefit from the inclusion of additional conserved antigens, which may contribute to broader coverage and enhanced efficacy, given their key roles in facilitating intestinal colonization and effective enterotoxin delivery. EatA and EtpA are widely conserved in diverse populations of ETEC, but their immunogenicity has only been studied in controlled human infection models and a population of children in Bangladesh. Here, we compared serologic responses to EatA, EtpA and heat-labile toxin in populations from endemic regions including Haitian children and subjects residing in Egypt, Cameroon, and Peru to US children and adults where ETEC infections are sporadic. We observed elevated IgG and IgA responses in individuals from endemic regions to each of the antigens studied. In a cohort of Haitian children, we observed increased immune responses following exposure to each of the profiled antigens. These findings reflect the wide distribution of ETEC infections across multiple endemic regions and support further evaluation of EatA and EtpA as candidate ETEC vaccine antigens.
Collapse
Affiliation(s)
- Frederick Matthew Kuhlmann
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA; (F.M.K.); (V.G.); (T.J.V.)
| | - Vadim Grigura
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA; (F.M.K.); (V.G.); (T.J.V.)
| | - Timothy J. Vickers
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA; (F.M.K.); (V.G.); (T.J.V.)
| | | | - Lora L. Iannotti
- Institute for Public Health, Brown School, Washington University in Saint Louis, Saint Louis, MO 63110, USA; (L.L.I.); (S.J.L.D.)
| | - Sherlie Jean Louis Dulience
- Institute for Public Health, Brown School, Washington University in Saint Louis, Saint Louis, MO 63110, USA; (L.L.I.); (S.J.L.D.)
| | - James M. Fleckenstein
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine in Saint Louis, Saint Louis, MO 63110, USA; (F.M.K.); (V.G.); (T.J.V.)
- Medicine Service, Infectious Diseases, Saint Louis VA Health Care System, St. Louis, MO 63110, USA
| |
Collapse
|
42
|
Berzosa M, Delgado-López A, Irache JM, Gamazo C. Optimization of Enterotoxigenic Escherichia coli (ETEC) Outer Membrane Vesicles Production and Isolation Method for Vaccination Purposes. Microorganisms 2023; 11:2088. [PMID: 37630648 PMCID: PMC10458947 DOI: 10.3390/microorganisms11082088] [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: 07/19/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 08/27/2023] Open
Abstract
The study addresses Enterotoxigenic Escherichia coli (ETEC), a significant concern in low-income countries. Despite its prevalence, there is no licensed vaccine against ETEC. Bacterial vesicle-based vaccines are promising due to their safety and diverse virulence factors. However, cost-effective production requires enhancing vesicle yield while considering altered properties due to isolation methods. The proposed method involves heat treatment and ultrafiltration to recover vesicles from bacterial cultures. Two vesicle types, collected from heat-treated (HT-OMV) or untreated (NT-OMV) cultures, were compared. Vesicles were isolated via ultrafiltration alone ("complete") or with ultracentrifugation ("sediment"). Preliminary findings suggest complete HT-OMV vesicles are suitable for an ETEC vaccine. They express important proteins (OmpA, OmpX, OmpW) and virulence factors (adhesin TibA). Sized optimally (50-200 nm) for mucosal vaccination, they activate macrophages, inducing marker expression (CD40, MHCII, CD80, CD86) and Th1/Th2 cytokine release (IL-6, MCP-1, TNF-α, IL12p70, IL-10). This study confirms non-toxicity in RAW 264.7 cells and the in vivo ability of complete HT-OMV to generate significant IgG2a/IgG1 serum antibodies. Results suggest promise for a cost-effective ETEC vaccine, requiring further research on in vivo toxicity, pathogen-specific antibody detection, and protective efficacy.
Collapse
Affiliation(s)
- Melibea Berzosa
- Department of Microbiology and Parasitology, Navarra Medical Research Institute (IdiSNA), University of Navarra, 31008 Pamplona, Spain
| | - Alberto Delgado-López
- Department of Microbiology and Parasitology, Navarra Medical Research Institute (IdiSNA), University of Navarra, 31008 Pamplona, Spain
| | - Juan Manuel Irache
- Department of Pharmacy and Pharmaceutical Technology, University of Navarra, 31008 Pamplona, Spain
| | - Carlos Gamazo
- Department of Microbiology and Parasitology, Navarra Medical Research Institute (IdiSNA), University of Navarra, 31008 Pamplona, Spain
| |
Collapse
|
43
|
Abstract
Pathogen genome sequencing has become a routine part of our response to active outbreaks of infectious disease and should be an important part of our preparations for future epidemics. In this Essay, we discuss the innovations that have enabled routine pathogen genome sequencing, as well as how genome sequences can be used to understand and control the spread of infectious disease. We also explore the impact of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) pandemic on the field of pathogen genomics and outline the challenges we must address to further improve the utility of pathogen genome sequencing in the future.
Collapse
Affiliation(s)
- Jason T Ladner
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, United States of America
- Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, United States of America
| | - Jason W Sahl
- The Pathogen and Microbiome Institute, Northern Arizona University, Flagstaff, Arizona, United States of America
- Department of Biological Sciences, Northern Arizona University, Flagstaff, Arizona, United States of America
| |
Collapse
|
44
|
Bajoria S, Antunez LR, Kumru OS, Klempner M, Wang Y, Cavacini LA, Joshi SB, Volkin DB. Formulation Studies to Develop Low-Cost, Orally-Delivered Secretory IgA Monoclonal Antibodies for Passive Immunization Against Enterotoxigenic Escherichia coli. J Pharm Sci 2023; 112:1832-1844. [PMID: 37040833 DOI: 10.1016/j.xphs.2023.04.005] [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: 01/12/2023] [Revised: 04/06/2023] [Accepted: 04/06/2023] [Indexed: 04/13/2023]
Abstract
Enterotoxigenic Escherichia coli (ETEC) is a common cause for diarrheal infections in children in low- and middle-income countries (LMICs). To date, no ETEC vaccine candidates have been approved. Passive immunization with low-cost, oral formulations of secretory IgA (sIgA) against ETEC is an alternative approach to protect high-risk populations in LMICs. Using a model sIgA monoclonal antibody (anti-LT sIgA2-mAb), the stability profiles of different formulations were assessed during storage and in in vitro digestion models (mimicking in vivo oral delivery). First, by employing various physicochemical techniques and a LT-antigen binding assay, three formulations with varying acid-neutralizing capacity (ANC) were evaluated to stabilize sIgA2-mAb during stress studies (freeze-thaw, agitation, elevated temperature) and during exposure to gastric phase digestion. Next, a low-volume, in vitro intestinal digestion model was developed to screen various additives to stabilize sIgA2-mAb in the intestinal phase. Finally, combinations of high ANC buffers and decoy proteins were assessed to collectively protect sIgA2-mAb during in vitro sequential (stomach to intestine) digestion. Based on the results, we demonstrate the feasibility of low-cost, 'single-vial', liquid formulations of sIgA-mAbs delivered orally after infant feeding for passive immunization, and we suggest future work based on a combination of in vitro and in vivo stability considerations.
Collapse
Affiliation(s)
- Sakshi Bajoria
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center (VAFC), University of Kansas, Lawrence, KS 66047, USA
| | - Lorena R Antunez
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center (VAFC), University of Kansas, Lawrence, KS 66047, USA
| | - Ozan S Kumru
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center (VAFC), University of Kansas, Lawrence, KS 66047, USA
| | - Mark Klempner
- MassBiologics of the University of Massachusetts Chan Medical School, Boston, MA 02126, USA
| | - Yang Wang
- MassBiologics of the University of Massachusetts Chan Medical School, Boston, MA 02126, USA
| | - Lisa A Cavacini
- MassBiologics of the University of Massachusetts Chan Medical School, Boston, MA 02126, USA
| | - Sangeeta B Joshi
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center (VAFC), University of Kansas, Lawrence, KS 66047, USA
| | - David B Volkin
- Department of Pharmaceutical Chemistry, Vaccine Analytics and Formulation Center (VAFC), University of Kansas, Lawrence, KS 66047, USA.
| |
Collapse
|
45
|
Park JY, Cho SH. Production of monoclonal antibody of heat-labile toxin A subunit to identify enterotoxigenic Escherichia coli by epitope mapping using synthetic peptides. Front Immunol 2023; 14:1152910. [PMID: 37275900 PMCID: PMC10232981 DOI: 10.3389/fimmu.2023.1152910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 05/05/2023] [Indexed: 06/07/2023] Open
Abstract
Background Enterotoxigenic Escherichia coli (ETEC) is a major cause of diarrhea through two enterotoxins, a heat-labile toxin and a heat-stable toxin. These toxins alter the cellular signaling pathways, ultimately triggering an increase in chloride secretion and watery diarrhea. Objective For the development of an ETEC vaccine, we attempted to construct a peptide-specific monoclonal antibody library against heat-labile enterotoxin A subunit (LT-A) by epitope mapping using synthetic peptides. Methods Sera produced by five mice immunized with recombinant LT-A protein were examined for specific recognition with synthetic 15-mer and 34-mer peptides of LT-A proteins using enzyme-linked immunosorbent assay. The analysis revealed that the synthetic peptides number 8, 16, 24, 33, 36, 38, and 39 reacted with an anti-LT-A polyclonal antibody. For the possible prediction of LT-A epitopes, each full-length protein sequence was subjected to BCPreds analysis and three-dimensional protein structure analysis. The data showed that three peptides (synthetic peptide numbers: 33, 36, and 38-39) have identical antigenic specificities with LT-A protein, suggesting the usefulness of these linear peptide epitopes. Results Based on these peptides, we produced monoclonal antibodies to improve the specificity of LT-A detection. Monoclonal antibodies produced from two peptides (numbers 33 and 36) showed affinity for an LT-A recombinant antigen. Moreover, peptide epitope prediction analysis showed that the sites of the three peptides were identical to those exhibiting actual antigenicity. Also, it was confirmed that the amino acid sequence that actually showed antigenicity was included in the peptide predicted only by ETEC-LT-A-33. Also, the specificity of the antibody for ETEC-LT-A-33 was validated using bacterial cells, and the neutralizing effect of the antibody was determined by assessing cytokine release in infected HCT-8 cells. Conclusion The monoclonal antibodies produced in this study are useful toolsfor vaccine production against ETEC and can be used to identify peptide antigencandidates.
Collapse
Affiliation(s)
- Jun-Young Park
- Division of Zoonotic and Vector Borne Disease Research, Center for Infectious Disease Research, Korea National Institute of Health, Cheongju, Republic of Korea
- Environmental Diseases Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Seung-Hak Cho
- Division of Zoonotic and Vector Borne Disease Research, Center for Infectious Disease Research, Korea National Institute of Health, Cheongju, Republic of Korea
| |
Collapse
|
46
|
Diaz JN, Iannotti LL, Louis Dulience SJ, Vie S, Jiang X, Grigura V, Boncy J, Pierre FJM, Kuhlmann FM. Prevalence of diarrheagenic Escherichia coli and impact on child health in Cap-Haitien, Haiti. PLOS GLOBAL PUBLIC HEALTH 2023; 3:e0001863. [PMID: 37145992 PMCID: PMC10162540 DOI: 10.1371/journal.pgph.0001863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/06/2023] [Indexed: 05/07/2023]
Abstract
BACKGROUND Diarrheagenic Escherichia coli (DEC) are common pathogens infecting children during their growth and development. Determining the epidemiology and the impact of DEC on child anthropometric measures informs prioritization of prevention efforts. These relationships were evaluated in a novel setting, Cap-Haitien, Haiti. METHODS We performed pre-specified secondary analysis of a case-control study of community-dwelling children, 6-36 months of age, enrolled 96 cases with diarrhea and 99 asymptomatic controls. Assessments were performed at enrollment and one month later at follow-up. Established endpoint PCR methodologies targeted DEC gDNA isolated from fecal swabs. The association between DEC and anthropometric z-scores at enrollment was determined using multivariate linear regression. Lastly, we assessed the association between specific biomarkers, choline and docosahexaenoic acid (DHA) and diarrheal burden. RESULTS Enterotoxigenic Escherichia coli (ETEC) was identified in 21.9% of cases vs. 16.1% of controls with heat-stable producing ETEC significantly associated with symptomatic disease. Enteroaggregative E. coli (EAEC) was found in 30.2% of cases vs. 27.3% of controls, and typical enteropathogenic E. coli in 6.3% vs. 4.0% of cases and controls, respectively. Multivariate linear regression, controlled for case or control status, demonstrated ETEC and EAEC were significantly associated with reduced weight-age z-score (WAZ) and height-age z-score (HAZ) after adjusting for confounders. An interaction between ETEC and EAEC was observed. Choline and DHA were not associated with diarrheal burden. CONCLUSIONS DEC are prevalent in north Haitian children. ETEC, EAEC, household environment, and diet are associated with unfavorable anthropometric measures, with possible synergistic interactions between ETEC and EAEC. Further studies with longer follow up may quantify the contribution of individual pathogens to adverse health outcomes.
Collapse
Affiliation(s)
- Jenna N. Diaz
- Department of Pediatrics, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Lora L. Iannotti
- Brown School, Institute for Public Health, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Sherlie Jean Louis Dulience
- Brown School, Institute for Public Health, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Sydney Vie
- Brown School, Institute for Public Health, Washington University in St. Louis, St. Louis, Missouri, United States of America
| | - Xuntian Jiang
- Department of Medicine, Diabetic Cardiovascular Disease Center, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Vadim Grigura
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, United States of America
| | - Jacques Boncy
- Laboratoire National de Santé Publique, Ministère de la Santé Publique et de la Population, Port-au-Prince, Haiti
| | - Francesca J. Marhône Pierre
- Unité de Coordination du Programme National d’Alimentation et de Nutrition, Ministère de la Santé Publique et de la Population, Port-au-Prince, Haiti
| | - F. Matthew Kuhlmann
- Laboratoire National de Santé Publique, Ministère de la Santé Publique et de la Population, Port-au-Prince, Haiti
| |
Collapse
|
47
|
Mubanga C, Simuyandi M, Mwape K, Chibesa K, Chisenga C, Chilyabanyama ON, Randall A, Liang X, Glashoff RH, Chilengi R. Use of an ETEC Proteome Microarray to Evaluate Cross-Reactivity of ETVAX ® Vaccine-Induced IgG Antibodies in Zambian Children. Vaccines (Basel) 2023; 11:vaccines11050939. [PMID: 37243042 DOI: 10.3390/vaccines11050939] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/02/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Developing a broadly protective vaccine covering most ETEC variants has been elusive. The most clinically advanced candidate yet is an oral inactivated ETEC vaccine (ETVAX®). We report on the use of a proteome microarray for the assessment of cross-reactivity of anti-ETVAX® IgG antibodies against over 4000 ETEC antigens and proteins. We evaluated 40 (pre-and post-vaccination) plasma samples from 20 Zambian children aged 10-23 months that participated in a phase 1 trial investigating the safety, tolerability, and immunogenicity of ETVAX® adjuvanted with dmLT. Pre-vaccination samples revealed high IgG responses to a variety of ETEC proteins including classical ETEC antigens (CFs and LT) and non-classical antigens. Post-vaccination reactivity to CFA/I, CS3, CS6, and LTB was stronger than baseline among the vaccinated compared to the placebo group. Interestingly, we noted significantly high post-vaccination responses to three non-vaccine ETEC proteins: CS4, CS14, and PCF071 (p = 0.043, p = 0.028, and p = 0.00039, respectively), suggestive of cross-reactive responses to CFA/I. However, similar responses were observed in the placebo group, indicating the need for larger studies. We conclude that the ETEC microarray is a useful tool for investigating antibody responses to numerous antigens, especially because it may not be practicable to include all antigens in a single vaccine.
Collapse
Affiliation(s)
- Cynthia Mubanga
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia
- Division of Medical Microbiology, Department of Pathology, Stellenbosch University & National Health Laboratory Service, Tygerberg Hospital Francie van Zijl Drive, P.O. Box 241, Cape Town 8000, South Africa
| | - Michelo Simuyandi
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia
| | - Kapambwe Mwape
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia
- Water and Health Research Center, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| | - Kennedy Chibesa
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia
- Division of Virology, School of Pathology, Faculty of Health Sciences, University of the Free State, 205 Nelson Mandela, P.O. Box 339, Bloemfontein 9300, South Africa
| | - Caroline Chisenga
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia
| | | | - Arlo Randall
- Antigen Discovery Inc., 1 Technology Dr., Suite E309, Irvine, CA 92618, USA
| | - Xiaowu Liang
- Antigen Discovery Inc., 1 Technology Dr., Suite E309, Irvine, CA 92618, USA
| | - Richard H Glashoff
- Division of Medical Microbiology, Department of Pathology, Stellenbosch University & National Health Laboratory Service, Tygerberg Hospital Francie van Zijl Drive, P.O. Box 241, Cape Town 8000, South Africa
| | - Roma Chilengi
- Enteric Disease and Vaccine Research Unit, Centre for Infectious Disease Research in Zambia, Lusaka P.O. Box 34681, Zambia
| |
Collapse
|
48
|
Walker RI, Bourgeois AL. Oral inactivated whole cell vaccine for mucosal immunization: ETVAX case study. Front Immunol 2023; 14:1125102. [PMID: 36936951 PMCID: PMC10018008 DOI: 10.3389/fimmu.2023.1125102] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/03/2023] [Indexed: 03/06/2023] Open
Abstract
Oral immunization is an effective strategy for inducing protective immunity against mucosal enteric pathogens. Although live-attenuated as well as subunit approaches have been explored for vaccination against enteric pathogens, inactivated whole bacterial cells may also be effective in introducing protective immunity. Successfully accomplishing this goal with inactivated whole bacterial cells will require that a complex antigenic repertoire be presented in controlled immunogenic amounts, in a safe and relatively simple and self-contained delivery format. The benefit from immunization with whole cell vaccines can be further enhanced through genetic engineering to over-express selected antigens and also by the use of mucosal adjuvants to direct a more robust immunologic response. These steps are being taken for the development of ETVAX, the most clinically advanced vaccine candidate against the major enteric pathogen, enterotoxigenic Escherichia coli (ETEC) with significant positive impact.
Collapse
Affiliation(s)
- Richard I. Walker
- Center for Vaccine Innovation and Access, PATH, Washington, DC, United States
| | | |
Collapse
|
49
|
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: 68] [Impact Index Per Article: 34.0] [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
|
50
|
Warfel K, Williams A, Wong DA, Sobol SE, Desai P, Li J, Chang YF, DeLisa MP, Karim AS, Jewett MC. A Low-Cost, Thermostable, Cell-Free Protein Synthesis Platform for On-Demand Production of Conjugate Vaccines. ACS Synth Biol 2023; 12:95-107. [PMID: 36548479 PMCID: PMC9872175 DOI: 10.1021/acssynbio.2c00392] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Indexed: 12/24/2022]
Abstract
Cell-free protein synthesis systems that can be lyophilized for long-term, non-refrigerated storage and transportation have the potential to enable decentralized biomanufacturing. However, increased thermostability and decreased reaction cost are necessary for further technology adoption. Here, we identify maltodextrin as an additive to cell-free reactions that can act as both a lyoprotectant to increase thermostability and a low-cost energy substrate. As a model, we apply optimized formulations to produce conjugate vaccines for ∼$0.50 per dose after storage at room temperature (∼22 °C) or 37 °C for up to 4 weeks, and ∼$1.00 per dose after storage at 50 °C for up to 4 weeks, with costs based on raw materials purchased at the laboratory scale. We show that these conjugate vaccines generate bactericidal antibodies against enterotoxigenic Escherichia coli (ETEC) O78 O-polysaccharide, a pathogen responsible for diarrheal disease, in immunized mice. We anticipate that our low-cost, thermostable cell-free glycoprotein synthesis system will enable new models of medicine biosynthesis and distribution that bypass cold-chain requirements.
Collapse
Affiliation(s)
- Katherine
F. Warfel
- Department
of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Technological Institute E136, Evanston, Illinois 60208, United States
- Chemistry
of Life Processes Institute, Northwestern
University, 2170 Campus
Drive, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, 2145 Sheridan Road, Technological
Institute E136, Evanston, Illinois 60208, United States
| | - Asher Williams
- Robert
Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853 United States
| | - Derek A. Wong
- Department
of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Technological Institute E136, Evanston, Illinois 60208, United States
- Chemistry
of Life Processes Institute, Northwestern
University, 2170 Campus
Drive, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, 2145 Sheridan Road, Technological
Institute E136, Evanston, Illinois 60208, United States
| | - Sarah E. Sobol
- Department
of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Technological Institute E136, Evanston, Illinois 60208, United States
- Chemistry
of Life Processes Institute, Northwestern
University, 2170 Campus
Drive, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, 2145 Sheridan Road, Technological
Institute E136, Evanston, Illinois 60208, United States
| | - Primit Desai
- Biochemistry,
Molecular & Cell Biology, Cornell University, Ithaca, New York 14853 United States
| | - Jie Li
- Department
of Population Medicine and Diagnostic Sciences, College of Veterinary
Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Yung-Fu Chang
- Department
of Population Medicine and Diagnostic Sciences, College of Veterinary
Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Matthew P. DeLisa
- Robert
Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York 14853 United States
- Biochemistry,
Molecular & Cell Biology, Cornell University, Ithaca, New York 14853 United States
- Cornell
Institute of Biotechnology, Cornell University, Ithaca, New York 14853 United States
| | - Ashty S. Karim
- Department
of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Technological Institute E136, Evanston, Illinois 60208, United States
- Chemistry
of Life Processes Institute, Northwestern
University, 2170 Campus
Drive, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, 2145 Sheridan Road, Technological
Institute E136, Evanston, Illinois 60208, United States
| | - Michael C. Jewett
- Department
of Chemical and Biological Engineering, Northwestern University, 2145 Sheridan Road, Technological Institute E136, Evanston, Illinois 60208, United States
- Chemistry
of Life Processes Institute, Northwestern
University, 2170 Campus
Drive, Evanston, Illinois 60208, United States
- Center
for Synthetic Biology, Northwestern University, 2145 Sheridan Road, Technological
Institute E136, Evanston, Illinois 60208, United States
- Robert
H. Lurie Comprehensive Cancer Center, Northwestern
University, 676 North
Saint Clair Street, Suite 1200, Chicago, Illinois 60611, United States
- Simpson
Querrey Institute, Northwestern University, 303 East Superior Street, Suite
11-131, Chicago, Illinois 60611, United States
| |
Collapse
|