1
|
Savitri CMA, Fauzia KA, Alfaray RI, Aftab H, Syam AF, Lubis M, Yamaoka Y, Miftahussurur M. Opportunities for Helicobacter pylori Eradication beyond Conventional Antibiotics. Microorganisms 2024; 12:1986. [PMID: 39458296 PMCID: PMC11509656 DOI: 10.3390/microorganisms12101986] [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: 07/31/2024] [Revised: 09/14/2024] [Accepted: 09/19/2024] [Indexed: 10/28/2024] Open
Abstract
Helicobacter pylori (H. pylori) is a bacterium known to be associated with a significant risk of gastric cancer in addition to chronic gastritis, peptic ulcer, and MALT lymphoma. Although only a small percentage of patients infected with H. pylori develop gastric cancer, Gastric cancer causes more than 750,000 deaths worldwide, with 90% of cases being caused by H. pylori. The eradication of this bacterium rests on multiple drug regimens as guided by various consensus. However, the efficacy of empirical therapy is decreasing due to antimicrobial resistance. In addition, biofilm formation complicates eradication. As the search for new antibiotics lags behind the bacterium's ability to mutate, studies have been directed toward finding new anti-H. pylori agents while also optimizing current drug functions. Targeting biofilm, repurposing outer membrane vesicles that were initially a virulence factor of the bacteria, phage therapy, probiotics, and the construction of nanoparticles might be able to complement or even be alternatives for H. pylori treatment. This review aims to present reports on various compounds, either new or combined with current antibiotics, and their pathways to counteract H. pylori resistance.
Collapse
Affiliation(s)
- Camilia Metadea Aji Savitri
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Oita, Japan; (C.M.A.S.); (R.I.A.)
- Helicobacter Pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya 60286, Indonesia;
| | - Kartika Afrida Fauzia
- Helicobacter Pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya 60286, Indonesia;
- Research Centre for Preclinical and Clinical Medicine, National Research and Innovation Agency, Cibinong Science Center, Bogor 16915, Indonesia
| | - Ricky Indra Alfaray
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Oita, Japan; (C.M.A.S.); (R.I.A.)
- Helicobacter Pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya 60286, Indonesia;
| | - Hafeza Aftab
- Department of Gastroenterology, Dhaka Medical College and Hospital, Dhaka 1000, Bangladesh;
| | - Ari Fahrial Syam
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia;
| | - Masrul Lubis
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sumatera Utara, Medan 20155, Indonesia;
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu 879-5593, Oita, Japan; (C.M.A.S.); (R.I.A.)
- Helicobacter Pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya 60286, Indonesia;
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, TX 77030, USA
- Division of Genome-Wide Microbiology, Research Center for Global and Local Infectious Diseases (RCGLID), Oita University, Yufu 879-5593, Oita, Japan
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine—Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya 60286, Indonesia
| | - Muhammad Miftahussurur
- Helicobacter Pylori and Microbiota Study Group, Institute of Tropical Disease, Universitas Airlangga, Surabaya 60286, Indonesia;
- Division of Gastroentero-Hepatology, Department of Internal Medicine, Faculty of Medicine—Dr. Soetomo Teaching Hospital, Universitas Airlangga, Surabaya 60286, Indonesia
| |
Collapse
|
2
|
Maity R, Zhang X, Liberati FR, Scribani Rossi C, Cutruzzolá F, Rinaldo S, Gaetani M, Aínsa JA, Sancho J. Merging multi-omics with proteome integral solubility alteration unveils antibiotic mode of action. eLife 2024; 13:RP96343. [PMID: 39329363 PMCID: PMC11434622 DOI: 10.7554/elife.96343] [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] [Indexed: 09/28/2024] Open
Abstract
Antimicrobial resistance is responsible for an alarming number of deaths, estimated at 5 million per year. To combat priority pathogens, like Helicobacter pylori, the development of novel therapies is of utmost importance. Understanding the molecular alterations induced by medications is critical for the design of multi-targeting treatments capable of eradicating the infection and mitigating its pathogenicity. However, the application of bulk omics approaches for unraveling drug molecular mechanisms of action is limited by their inability to discriminate between target-specific modifications and off-target effects. This study introduces a multi-omics method to overcome the existing limitation. For the first time, the Proteome Integral Solubility Alteration (PISA) assay is utilized in bacteria in the PISA-Express format to link proteome solubility with different and potentially immediate responses to drug treatment, enabling us the resolution to understand target-specific modifications and off-target effects. This study introduces a comprehensive method for understanding drug mechanisms and optimizing the development of multi-targeting antimicrobial therapies.
Collapse
Affiliation(s)
- Ritwik Maity
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Unit GBsC-CSIC, University of ZaragozaZaragozaSpain
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of ZaragozaZaragozaSpain
- Aragon Health Research Institute (IIS Aragón)ZaragozaSpain
| | - Xuepei Zhang
- Department of Medical Biochemistry and Biophysics, Karolinska InstitutetStockholmSweden
- Chemical Proteomics Unit, Science for Life Laboratory (SciLifeLab)StockholmSweden
- Chemical Proteomics, Swedish National Infrastructure for Biological Mass Spectrometry (BioMS)StockholmSweden
| | | | - Chiara Scribani Rossi
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of RomeRomeItaly
| | - Francesca Cutruzzolá
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of RomeRomeItaly
| | - Serena Rinaldo
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of RomeRomeItaly
| | - Massimiliano Gaetani
- Department of Medical Biochemistry and Biophysics, Karolinska InstitutetStockholmSweden
- Chemical Proteomics Unit, Science for Life Laboratory (SciLifeLab)StockholmSweden
- Chemical Proteomics, Swedish National Infrastructure for Biological Mass Spectrometry (BioMS)StockholmSweden
| | - José Antonio Aínsa
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Unit GBsC-CSIC, University of ZaragozaZaragozaSpain
- Aragon Health Research Institute (IIS Aragón)ZaragozaSpain
- Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Faculty of Medicine, University of ZaragozaZaragozaSpain
- CIBER de Enfermedades Respiratorias—CIBERES, Instituto de Salud Carlos IIIMadridSpain
| | - Javier Sancho
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Unit GBsC-CSIC, University of ZaragozaZaragozaSpain
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of ZaragozaZaragozaSpain
- Aragon Health Research Institute (IIS Aragón)ZaragozaSpain
| |
Collapse
|
3
|
Umar Z, Tang JW, Marshall BJ, Tay ACY, Wang L. Rapid diagnosis and precision treatment of Helicobacter pylori infection in clinical settings. Crit Rev Microbiol 2024:1-30. [PMID: 38910506 DOI: 10.1080/1040841x.2024.2364194] [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: 02/28/2024] [Accepted: 05/25/2024] [Indexed: 06/25/2024]
Abstract
Helicobacter pylori is a gram-negative bacterium that colonizes the stomach of approximately half of the worldwide population, with higher prevalence in densely populated areas like Asia, the Caribbean, Latin America, and Africa. H. pylori infections range from asymptomatic cases to potentially fatal diseases, including peptic ulcers, chronic gastritis, and stomach adenocarcinoma. The management of these conditions has become more difficult due to the rising prevalence of drug-resistant H. pylori infections, which ultimately lead to gastric cancer and mucosa-associated lymphoid tissue (MALT) lymphoma. In 1994, the International Agency for Research on Cancer (IARC) categorized H. pylori as a Group I carcinogen, contributing to approximately 780,000 cancer cases annually. Antibiotic resistance against drugs used to treat H. pylori infections ranges between 15% and 50% worldwide, with Asian countries having exceptionally high rates. This review systematically examines the impacts of H. pylori infection, the increasing prevalence of antibiotic resistance, and the urgent need for accurate diagnosis and precision treatment. The present status of precision treatment strategies and prospective approaches for eradicating infections caused by antibiotic-resistant H. pylori will also be evaluated.
Collapse
Affiliation(s)
- Zeeshan Umar
- Marshall Laboratory of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, Guangdong Province, China
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
| | - Jia-Wei Tang
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- The Marshall Centre for Infectious Diseases Research and Training, The University of Western Australia, Crawley, Western Australia, China
| | - Barry J Marshall
- Marshall Laboratory of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, Guangdong Province, China
- The Marshall Centre for Infectious Diseases Research and Training, The University of Western Australia, Crawley, Western Australia, China
- Marshall International Digestive Diseases Hospital, Zhengzhou University, Zhengzhou, Henan Province, China
- Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Alfred Chin Yen Tay
- Marshall Laboratory of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen, Guangdong Province, China
- The Marshall Centre for Infectious Diseases Research and Training, The University of Western Australia, Crawley, Western Australia, China
- Marshall International Digestive Diseases Hospital, Zhengzhou University, Zhengzhou, Henan Province, China
- Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Liang Wang
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong Province, China
- Division of Microbiology and Immunology, School of Biomedical Sciences, The University of Western Australia, Crawley, Western Australia, China
- Center for Precision Health, School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, China
- School of Agriculture and Food Sustainability, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
4
|
Galano-Frutos JJ, Maity R, Iguarbe V, Aínsa JA, Velázquez-Campoy A, Schaible UE, Mamat U, Sancho J. L-Thyroxine and L-thyroxine-based antimicrobials against Streptococcus pneumoniae and other Gram-positive bacteria. Heliyon 2024; 10:e27982. [PMID: 38689973 PMCID: PMC11059415 DOI: 10.1016/j.heliyon.2024.e27982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 05/02/2024] Open
Abstract
Objectives The rise of antibiotic-resistant Streptococcus pneumoniae (Sp) poses a significant global health threat, urging the quest for novel antimicrobial solutions. We have discovered that the human hormone l-thyroxine has antibacterial properties. In order to explore its drugability we perform here the characterization of a series of l-thyroxine analogues and describe the structural determinants influencing their antibacterial efficacy. Method We performed a high-throughput screening of a library of compounds approved for use in humans, complemented with ITC assays on purified Sp-flavodoxin, to pinpoint molecules binding to this protein. Antimicrobial in vitro susceptibility assays of the hit compound (l-thyroxine) as well as of 13 l-thyroxine analogues were done against a panel of Gram-positive and Gram-negative bacteria. Toxicity of compounds on HepG2 cells was also assessed. A combined structure-activity and computational docking analysis was carried out to uncover functional groups crucial for the antimicrobial potency of these compounds. Results Human l-thyroxine binds to Sp-flavodoxin, forming a 1:1 complex of low micromolar Kd. While l-thyroxine specifically inhibited Sp growth, some derivatives displayed activity against other Gram-positive bacteria like Staphylococcus aureus and Enterococcus faecalis, while remaining inactive against Gram-negative pathogens. Neither l-thyroxine nor some selected derivatives exhibited toxicity to HepG2 cells. Conclusions l-thyroxine derivatives targeting bacterial flavodoxins represent a new and promising class of antimicrobials.
Collapse
Affiliation(s)
- Juan José Galano-Frutos
- Institute of Chemical Sciences and Technologies "Giulio Natta" (SCITEC) - CNR, Largo Francesco Vito 1, 00168, Rome, Italy
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, Zaragoza 50018, Spain
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, University of Zaragoza, Zaragoza 50009, Spain
| | - Ritwik Maity
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, Zaragoza 50018, Spain
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, University of Zaragoza, Zaragoza 50009, Spain
| | - Verónica Iguarbe
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, Zaragoza 50018, Spain
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, University of Zaragoza, Zaragoza 50009, Spain
| | - José Antonio Aínsa
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, Zaragoza 50018, Spain
- Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Facultad de Medicina, University of Zaragoza, Zaragoza 50009, Spain
- CIBER de Enfermedades Respiratorias–CIBERES, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Adrián Velázquez-Campoy
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, Zaragoza 50018, Spain
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, University of Zaragoza, Zaragoza 50009, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza 50009, Spain
- CIBER de Enfermedades Hepáticas y Digestivas CIBERehd, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Ulrich E. Schaible
- Cellular Microbiology, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, & Leibniz Research Alliance INFECTIONS, Borstel, Germany
- Biochemical Microbiology & Immunochemistry, University of Lübeck, Lübeck, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg-Lübeck-Borstel-Riems, Borstel Germany
| | - Uwe Mamat
- Cellular Microbiology, Priority Research Area Infections, Research Center Borstel, Leibniz Lung Center, & Leibniz Research Alliance INFECTIONS, Borstel, Germany
| | - Javier Sancho
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, Zaragoza 50018, Spain
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, University of Zaragoza, Zaragoza 50009, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza 50009, Spain
| |
Collapse
|
5
|
Beyria L, Gourbeyre O, Salillas S, Mahía A, Díaz de Villegas MD, Aínsa JA, Sancho J, Bousquet-Mélou A, Ferran AA. Antimicrobial combinations against Helicobacter pylori including benzoxadiazol-based flavodoxin inhibitors: in vitro characterization. Microbiol Spectr 2024; 12:e0262323. [PMID: 38084974 PMCID: PMC10783109 DOI: 10.1128/spectrum.02623-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 11/05/2023] [Indexed: 01/13/2024] Open
Abstract
IMPORTANCE The antimicrobial resistance of Helicobacter pylori (Hp) currently poses a threat to available treatment regimens. Developing antimicrobial drugs targeting new bacterial targets is crucial, and one such class of drugs includes Hp-flavodoxin (Hp-fld) inhibitors that target an essential metabolic pathway in Hp. Our study demonstrated that combining these new drugs with conventional antibiotics used for Hp infection treatment prevented the regrowth observed with drugs used alone. Hp-fld inhibitors show promise as new drugs to be incorporated into the treatment of Hp infection, potentially reducing the development of resistance and shortening the treatment duration.
Collapse
Affiliation(s)
- Lilha Beyria
- INTHERES, Université de Toulouse, INRAE, ENVT, Toulouse, France
| | | | - Sandra Salillas
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, Zaragoza, Spain
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, University of Zaragoza, Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - Alejandro Mahía
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, Zaragoza, Spain
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, University of Zaragoza, Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
| | - María Dolores Díaz de Villegas
- CSIC—Departamento de Química Orgánica, Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), University of Zaragoza, Zaragoza, Spain
| | - José Antonio Aínsa
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
- Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Facultad de Medicina, University of Zaragoza, Zaragoza, Spain
- CIBER de Enfermedades Respiratorias–CIBERES, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Sancho
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, Zaragoza, Spain
- Departamento de Bioquímica y Biología Molecular y Celular, Facultad de Ciencias, University of Zaragoza, Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), Zaragoza, Spain
| | | | - Aude A. Ferran
- INTHERES, Université de Toulouse, INRAE, ENVT, Toulouse, France
| |
Collapse
|
6
|
Li F, Zhao Z, Chen W, Liu R, Lu H, Dong Y, Yang Q, Zhang J. Design, Synthesis, and Biological Investigations of Novel Carbamoylguanidinyl Nitrobenzoxadiazoles against Chitinolytic Enzymes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:18333-18344. [PMID: 37967522 DOI: 10.1021/acs.jafc.3c06157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2023]
Abstract
Chitinase has been identified as an important target for insecticides. In this study, a series of novel chitinase inhibitors was designed and synthesized with nitrobenzoxadiazoles. Compound 8d, which contains the N-methylcarbamoylguanidinyl, exhibited high enzyme inhibitory activity and achieved nanomolar inhibition against OfChtI (IC50 = 12.3 nM). Delightfully, it was also found to possess significant inhibitory activity against OfHex1 (IC50 = 1.76 μM). The computational simulation results indicated that compound 8d interacted with OfChtI and OfHex1 in similar modes through hydrogen bonds and hydrophobic and π-π interactions. Insecticidal activity studies revealed that compound 8d showed high mortality against the Lepidoptera Plutella xylostella (mortality rate = 81%) at 200 mg/L. Toxicity studies indicated that compound 8d exhibited negligible toxicity to the natural enemy Trichogramma ostriniae. These results indicate that compound 8d may be a promising candidate for the development of environmentally friendly chitinase inhibitors. Moreover, this study provides a new angle for the design of innovative inhibitors of chitinolytic enzymes.
Collapse
Affiliation(s)
- Fang Li
- Department of Applied Chemistry, China Agricultural University, Beijing 100193, China
| | - Zhixiang Zhao
- Department of Applied Chemistry, China Agricultural University, Beijing 100193, China
| | - Wei Chen
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection and Shenzhen Agricultural Genome Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100193, People's Republic of China
| | - Ruiyuan Liu
- Department of Applied Chemistry, China Agricultural University, Beijing 100193, China
| | - Huizhe Lu
- Department of Applied Chemistry, China Agricultural University, Beijing 100193, China
| | - Yanhong Dong
- Department of Applied Chemistry, China Agricultural University, Beijing 100193, China
| | - Qing Yang
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Institute of Plant Protection and Shenzhen Agricultural Genome Research Institute, Chinese Academy of Agricultural Sciences, Beijing 100193, People's Republic of China
| | - Jianjun Zhang
- Department of Applied Chemistry, China Agricultural University, Beijing 100193, China
| |
Collapse
|
7
|
Tamulienė J, Valiauga B, Čėnas N. Modified quantum mechanical approach for the estimation of single-electron reduction potential of nitroaromatic compounds in aqueous medium. Free Radic Res 2023:1-8. [PMID: 37171206 DOI: 10.1080/10715762.2023.2213821] [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: 05/13/2023]
Abstract
The midpoint single-electron reduction potential of nitroaromatic compounds in aqueous medium at pH 7.0 (potential of ArNO2/ArNO2·- couple, Em7) frequently determines their therapeutic and/or toxic properties. However, its estimation remains a complex problem. We propose a modified method of Em7 estimation by quantum mechanical calculations, based on the use of the dielectric continuum model together with a certain number of H2O molecules at the vicinity of nitro group. The optimal number of H2O molecules corresponds to a minimal difference between the experimentally determined and calculated values of Em7, and/or the most negative value of calculated Em7. This enabled us to calculate the Em7 values for a number of ArNO2 (n = 19) with the average deviation of 0.027 V from the experimentally determined ones. Apart from nitrobenzene derivatives, the application of this approach for the representatives of nitropyridines, nitrofurans, nitrothiophenes, and nitrothiazoles was demonstrated. In this case, nitroimidazole derivatives are an exception, evidently due to a strong proton accepting properties of N3 atom of their free radicals.
Collapse
Affiliation(s)
- Jelena Tamulienė
- Vilnius University, Faculty of Physics, Institute of Theoretical Physics and Astronomy, Vilnius, Lithuania
| | | | - Narimantas Čėnas
- Institute of Biochemistry of Vilnius University, Vilnius, Lithuania
| |
Collapse
|
8
|
Srisuphanunt M, Wilairatana P, Kooltheat N, Duangchan T, Katzenmeier G, Rose JB. Molecular Mechanisms of Antibiotic Resistance and Novel Treatment Strategies for Helicobacter pylori Infections. Trop Med Infect Dis 2023; 8:163. [PMID: 36977164 PMCID: PMC10057134 DOI: 10.3390/tropicalmed8030163] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/06/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Helicobacter pylori infects approximately 50% of the world's population and is considered the major etiological agent of severe gastric diseases, such as peptic ulcers and gastric carcinoma. Increasing resistance to standard antibiotics has now led to an ever-decreasing efficacy of eradication therapies and the development of novel and improved regimens for treatment is urgently required. Substantial progress has been made over the past few years in the identification of molecular mechanisms which are conducive to resistant phenotypes as well as for efficient strategies to counteract strain resistance and to avoid the use of ineffective antibiotics. These involve molecular testing methods, improved salvage therapies, and the discovery of novel and potent antimicrobial compounds. High rates of prevalence and gastric cancer are currently observed in Asian countries, including Japan, China, Korea, and Taiwan, where concomitantly intensive research efforts were initiated to explore advanced eradication regimens aimed at reducing the risk of gastric cancer. In this review, we present an overview of the known molecular mechanisms of antibiotic resistance and discuss recent intervention strategies for H. pylori diseases, with a view of the research progress in Asian countries.
Collapse
Affiliation(s)
- Mayuna Srisuphanunt
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Excellent Center for Dengue and Community Public Health, School of Public Health, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Nateelak Kooltheat
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Hematology and Transfusion Science Research Center, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Thitinat Duangchan
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
- Hematology and Transfusion Science Research Center, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Gerd Katzenmeier
- Akkhraratchakumari Veterinary College, Walailak University, Nakhon Si Thammarat 80160, Thailand
| | - Joan B. Rose
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI 48823, USA
| |
Collapse
|
9
|
Targeting Helicobacter pylori for antibacterial drug discovery with novel therapeutics. Curr Opin Microbiol 2022; 70:102203. [PMID: 36156373 DOI: 10.1016/j.mib.2022.102203] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/11/2022] [Accepted: 08/16/2022] [Indexed: 01/25/2023]
Abstract
Helicobacter pylori is an important human pathogen with increasing antimicrobial resistance to standard-of-care antibiotics. Treatment generally includes a combination of classical broad-spectrum antibiotics and a proton-pump inhibitor, which often leads to perturbation of the gut microbiome and the potential for the development of antibiotic resistance. In this review, we examine reports, primarily from the past decade, on the discovery of new anti-H. pylori therapeutics, including approaches to develop narrow-spectrum and mechanistically unique antibiotics to treat these infections in their gastric niche. Compound series that target urease, respiratory complex I, and menaquinone biosynthesis are discussed in this context, along with bivalent antibiotic approaches that suppress resistance development. With increases in the understanding of the unique physiology of H. pylori and technological advances in the field of antibacterial drug discovery, there is a clear promise that novel therapeutics can be developed to effectively treat H. pylori infections.
Collapse
|
10
|
González A, Casado J, Gündüz MG, Santos B, Velázquez-Campoy A, Sarasa-Buisan C, Fillat MF, Montes M, Piazuelo E, Lanas Á. 1,4-Dihydropyridine as a Promising Scaffold for Novel Antimicrobials Against Helicobacter pylori. Front Microbiol 2022; 13:874709. [PMID: 35694298 PMCID: PMC9174938 DOI: 10.3389/fmicb.2022.874709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 05/03/2022] [Indexed: 12/19/2022] Open
Abstract
The increasing occurrence of multidrug-resistant strains of the gastric carcinogenic bacterium Helicobacter pylori threatens the efficacy of current eradication therapies. In a previous work, we found that several 1,4-dihydropyridine (DHP)-based antihypertensive drugs exhibited strong bactericidal activities against H. pylori by targeting the essential response regulator HsrA. To further evaluate the potential of 1,4-DHP as a scaffold for novel antimicrobials against H. pylori, we determined the antibacterial effects of 12 novel DHP derivatives that have previously failed to effectively block L- and T-type calcium channels. Six of these molecules exhibited potent antimicrobial activities (MIC ≤ 8 mg/L) against three different antibiotic-resistant strains of H. pylori, while at least one compound resulted as effective as metronidazole. Such antimicrobial actions appeared to be specific against Epsilonproteobacteria, since no deleterious effects were appreciated on Escherichia coli and Staphylococcus epidermidis. The new bactericidal DHP derivatives targeted the H. pylori regulator HsrA and inhibited its DNA binding activity according to both in vitro and in vivo analyses. Molecular docking predicted a potential druggable binding pocket in HsrA, which could open the door to structure-based design of novel anti-H. pylori drugs.
Collapse
Affiliation(s)
- Andrés González
- Group of Translational Research in Digestive Diseases, Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
- Department of Medicine, Psychiatry and Dermatology, University of Zaragoza, Zaragoza, Spain
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Zaragoza, Spain
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBERehd), Madrid, Spain
| | - Javier Casado
- Group of Translational Research in Digestive Diseases, Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
- Department of Biochemistry and Molecular and Cellular Biology, University of Zaragoza, Zaragoza, Spain
| | - Miyase Gözde Gündüz
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Brisa Santos
- Department of Biochemistry and Molecular and Cellular Biology, University of Zaragoza, Zaragoza, Spain
| | - Adrián Velázquez-Campoy
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Zaragoza, Spain
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBERehd), Madrid, Spain
- Department of Biochemistry and Molecular and Cellular Biology, University of Zaragoza, Zaragoza, Spain
- Fundación Agencia Aragonesa para la Investigación y el Desarrollo (ARAID), Zaragoza, Spain
| | - Cristina Sarasa-Buisan
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Zaragoza, Spain
- Department of Biochemistry and Molecular and Cellular Biology, University of Zaragoza, Zaragoza, Spain
| | - María F. Fillat
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Zaragoza, Spain
- Department of Biochemistry and Molecular and Cellular Biology, University of Zaragoza, Zaragoza, Spain
| | - Milagrosa Montes
- Department of Microbiology, Donostia University Hospital-Biodonostia Health Research Institute, San Sebastian, Spain
- Biomedical Research Networking Center in Respiratory Diseases (CIBERES), Madrid, Spain
| | - Elena Piazuelo
- Group of Translational Research in Digestive Diseases, Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBERehd), Madrid, Spain
- Aragón Health Sciences Institute (IACS), Zaragoza, Spain
| | - Ángel Lanas
- Group of Translational Research in Digestive Diseases, Institute for Health Research Aragón (IIS Aragón), Zaragoza, Spain
- Department of Medicine, Psychiatry and Dermatology, University of Zaragoza, Zaragoza, Spain
- Biomedical Research Networking Center in Hepatic and Digestive Diseases (CIBERehd), Madrid, Spain
- Digestive Diseases Service, University Clinic Hospital Lozano Blesa, Zaragoza, Spain
| |
Collapse
|
11
|
Xu M, Zhou J, Heng D, Su X, Onakpa MM, Bai Y, Duan JA, Che CT, Bi H, Zhao M. Quinone Derivatives as Promising Anti- Helicobacter pylori Agents from Aerial Parts of Mitracarpus hirtus. JOURNAL OF NATURAL PRODUCTS 2022; 85:1029-1038. [PMID: 35412828 DOI: 10.1021/acs.jnatprod.1c01163] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Seven new naphthoquinone diglycosides (1-7), three new anthraquinones (8-10), and eight known analogues were obtained from the aerial parts of Mitracarpus hirtus collected from West Africa in a bioassay-guided phytochemical investigation. All isolated compounds were elucidated by comparison with the literature and interpretation of spectroscopic data, and the absolute configurations of the new naphthoquinone diglycosides (1-10) were confirmed by chemical methods and ECD calculations. Notably, compound 1 was found to be the first naphthoquinone diglycoside containing carboxylic acid and isopentenyl side chains isolated from a species in the genus Mitracarpus. Compounds 6-18 showed antibacterial activity against multiple Helicobacter pylori strains with MIC values ranging from 0.0625 to 64 μg/mL. Particularly, 1-hydroxybenzoisochromanquinone (17) and benzo[g]isoquinoline-5,10-dione (18), with MIC values of 0.0625 and 0.125 μg/mL, displayed 32-512-fold higher potencies than a positive control, metronidazole. Compound 18 also demonstrated high antibiofilm activity and killed biofilm-encased Helicobacter pylori cells more effectively than metronidazole.
Collapse
Affiliation(s)
- Mingming Xu
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Junfei Zhou
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Ding Heng
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Helicobacter pylori Research Centre, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Xiaorong Su
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, People's Republic of China
| | - Monday M Onakpa
- Department of Veterinary Pharmacology and Toxicology, University of Abuja, Abuja 920001, Nigeria
| | - Yuefan Bai
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Helicobacter pylori Research Centre, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
| | - Chun-Tao Che
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Hongkai Bi
- Department of Pathogen Biology, Jiangsu Key Laboratory of Pathogen Biology, Helicobacter pylori Research Centre, Nanjing Medical University, Nanjing 211166, People's Republic of China
| | - Ming Zhao
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, People's Republic of China
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| |
Collapse
|
12
|
Salillas S, Galano-Frutos JJ, Mahía A, Maity R, Conde-Giménez M, Anoz-Carbonell E, Berlamont H, Velazquez-Campoy A, Touati E, Mamat U, Schaible UE, Gálvez JA, Díaz-de-Villegas MD, Haesebrouck F, Aínsa JA, Sancho J. Selective Targeting of Human and Animal Pathogens of the Helicobacter Genus by Flavodoxin Inhibitors: Efficacy, Synergy, Resistance and Mechanistic Studies. Int J Mol Sci 2021; 22:ijms221810137. [PMID: 34576300 PMCID: PMC8467567 DOI: 10.3390/ijms221810137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 02/08/2023] Open
Abstract
Antimicrobial resistant (AMR) bacteria constitute a global health concern. Helicobacter pylori is a Gram-negative bacterium that infects about half of the human population and is a major cause of peptic ulcer disease and gastric cancer. Increasing resistance to triple and quadruple H. pylori eradication therapies poses great challenges and urges the development of novel, ideally narrow spectrum, antimicrobials targeting H. pylori. Here, we describe the antimicrobial spectrum of a family of nitrobenzoxadiazol-based antimicrobials initially discovered as inhibitors of flavodoxin: an essential H. pylori protein. Two groups of inhibitors are described. One group is formed by narrow-spectrum compounds, highly specific for H. pylori, but ineffective against enterohepatic Helicobacter species and other Gram-negative or Gram-positive bacteria. The second group includes extended-spectrum antimicrobials additionally targeting Gram-positive bacteria, the Gram-negative Campylobacter jejuni, and most Helicobacter species, but not affecting other Gram-negative pathogens. To identify the binding site of the inhibitors in the flavodoxin structure, several H. pylori-flavodoxin variants have been engineered and tested using isothermal titration calorimetry. An initial study of the inhibitors capacity to generate resistances and of their synergism with antimicrobials commonly used in H. pylori eradication therapies is described. The narrow-spectrum inhibitors, which are expected to affect the microbiota less dramatically than current antimicrobial drugs, offer an opportunity to develop new and specific H. pylori eradication combinations to deal with AMR in H. pylori. On the other hand, the extended-spectrum inhibitors constitute a new family of promising antimicrobials, with a potential use against AMR Gram-positive bacterial pathogens.
Collapse
Affiliation(s)
- Sandra Salillas
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - Juan José Galano-Frutos
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - Alejandro Mahía
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - Ritwik Maity
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - María Conde-Giménez
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - Ernesto Anoz-Carbonell
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - Helena Berlamont
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B9820 Merelbeke, Belgium; (H.B.); (F.H.)
| | - Adrian Velazquez-Campoy
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- ARAID Foundation, Government of Aragon, 50018 Zaragoza, Spain
- CIBER de Enfermedades Hepáticas y Digestivas CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Eliette Touati
- Unit of Helicobacter Pathogenesis, CNRS UMR2001, Department of Microbiology, Institut Pasteur, 25-28 Rue du Dr. Roux, 75724 Paris, France;
| | - Uwe Mamat
- Cellular Microbiology, Program Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany; (U.M.); (U.E.S.)
| | - Ulrich E. Schaible
- Cellular Microbiology, Program Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany; (U.M.); (U.E.S.)
| | - José A. Gálvez
- Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC—Departamento de Química Orgánica, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain; (J.A.G.); (M.D.D.-d.-V.)
| | - María D. Díaz-de-Villegas
- Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC—Departamento de Química Orgánica, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain; (J.A.G.); (M.D.D.-d.-V.)
| | - Freddy Haesebrouck
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B9820 Merelbeke, Belgium; (H.B.); (F.H.)
| | - José A. Aínsa
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
- CIBER de Enfermedades Respiratorias—CIBERES, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Sancho
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- Correspondence:
| |
Collapse
|
13
|
Čėnas N, Nemeikaitė-Čėnienė A, Kosychova L. Single- and Two-Electron Reduction of Nitroaromatic Compounds by Flavoenzymes: Mechanisms and Implications for Cytotoxicity. Int J Mol Sci 2021; 22:ijms22168534. [PMID: 34445240 PMCID: PMC8395237 DOI: 10.3390/ijms22168534] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/30/2021] [Accepted: 08/04/2021] [Indexed: 12/14/2022] Open
Abstract
Nitroaromatic compounds (ArNO2) maintain their importance in relation to industrial processes, environmental pollution, and pharmaceutical application. The manifestation of toxicity/therapeutic action of nitroaromatics may involve their single- or two-electron reduction performed by various flavoenzymes and/or their physiological redox partners, metalloproteins. The pivotal and still incompletely resolved questions in this area are the identification and characterization of the specific enzymes that are involved in the bioreduction of ArNO2 and the establishment of their contribution to cytotoxic/therapeutic action of nitroaromatics. This review addresses the following topics: (i) the intrinsic redox properties of ArNO2, in particular, the energetics of their single- and two-electron reduction in aqueous medium; (ii) the mechanisms and structure-activity relationships of reduction in ArNO2 by flavoenzymes of different groups, dehydrogenases-electrontransferases (NADPH:cytochrome P-450 reductase, ferredoxin:NADP(H) oxidoreductase and their analogs), mammalian NAD(P)H:quinone oxidoreductase, bacterial nitroreductases, and disulfide reductases of different origin (glutathione, trypanothione, and thioredoxin reductases, lipoamide dehydrogenase), and (iii) the relationships between the enzymatic reactivity of compounds and their activity in mammalian cells, bacteria, and parasites.
Collapse
Affiliation(s)
- Narimantas Čėnas
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania;
- Correspondence: ; Tel.: +370-5-223-4392
| | - Aušra Nemeikaitė-Čėnienė
- State Research Institute Center for Innovative Medicine, Santariškių St. 5, LT-08406 Vilnius, Lithuania;
| | - Lidija Kosychova
- Institute of Biochemistry of Vilnius University, Saulėtekio 7, LT-10257 Vilnius, Lithuania;
| |
Collapse
|
14
|
Ghobadi E, Ghanbarimasir Z, Emami S. A review on the structures and biological activities of anti-Helicobacter pylori agents. Eur J Med Chem 2021; 223:113669. [PMID: 34218084 DOI: 10.1016/j.ejmech.2021.113669] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/11/2021] [Accepted: 06/21/2021] [Indexed: 12/19/2022]
Abstract
Helicobacter pylori is one of the main causal risk factor in the generation of chronic gastritis, gastroduodenal ulcers and gastric carcinoma. Thus, the eradication of H. pylori infection is an important way for preventing and managing the gastric diseases. Multiple-therapy with several antibacterial agents is used for the eradication of H. pylori infections; however the increase of resistance to H. pylori strains has resulted in unsatisfactory eradication and unsuccessful treatment. Furthermore, the combination therapy with high dosing leads to the disruption of intestinal microbial flora and undesired side effects. Therefore, the search for new therapeutic agents with high selectivity against H. pylori is a field of current interest. In recent years, diverse compounds originating from natural sources or synthetic drug design programs were evaluated and tried to optimize for applying against H. pylori. In this review, we have described various classes of anti-H. pylori compounds, their structure-activity relationship studies, and mechanism of actions, which could be useful for the development of new drugs for the treatment of H. pylori infections.
Collapse
Affiliation(s)
- Elham Ghobadi
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Zahra Ghanbarimasir
- Department of Organic Chemistry, Faculty of Chemistry, University of Mazandaran, Babolsar, Iran
| | - Saeed Emami
- Department of Medicinal Chemistry and Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
15
|
Abstract
Incorporation of heterocycles into drug molecules can enhance physical properties and biological activity. A variety of heterocyclic groups is available to medicinal chemists, many of which have been reviewed in detail elsewhere. Oxadiazoles are a class of heterocycle containing one oxygen and two nitrogen atoms, available in three isomeric forms. While the 1,2,4- and 1,3,4-oxadiazoles have seen widespread application in medicinal chemistry, 1,2,5-oxadiazoles (furazans) are less common. This Review provides a summary of the application of furazan-containing molecules in medicinal chemistry and drug development programs from analysis of both patent and academic literature. Emphasis is placed on programs that reached clinical or preclinical stages of development. The examples provided herein describe the pharmacology and biological activity of furazan derivatives with comparative data provided where possible for other heterocyclic groups and pharmacophores commonly used in medicinal chemistry.
Collapse
Affiliation(s)
| | | | - Donald F Weaver
- Department of Fundamental Neurobiology, Krembil Research Institute, Toronto, Ontario M5T 0S8, Canada.,Department of Chemistry, University of Toronto, Toronto, Ontario M5S 3H6, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario M5S 3H2, Canada
| | - Mark A Reed
- Treventis Corporation, Toronto, Ontario M5T 0S8, Canada.,Department of Fundamental Neurobiology, Krembil Research Institute, Toronto, Ontario M5T 0S8, Canada
| |
Collapse
|
16
|
Zhu H, Wang C, Zong L. Progress on Biological Activity Study and Enantioselective Synthesis of Sulfoxides. CHINESE J ORG CHEM 2021. [DOI: 10.6023/cjoc202103046] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
17
|
Xu MM, Zhou J, Zeng L, Xu J, Onakpa MM, Duan JA, Che CT, Bi H, Zhao M. Pimarane-derived diterpenoids with anti- Helicobacter pylori activity from the tuber of Icacina trichantha. Org Chem Front 2021. [DOI: 10.1039/d1qo00374g] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Two novel diterpenoids and ten known analogs were obtained from the tuber of Icacina trichantha. All compounds exhibited antibacterial activity against Helicobacter pylori strains with MIC values ranging from 8 to 64 μg mL−1.
Collapse
Affiliation(s)
- Ming-Ming Xu
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization
- School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing 210023
- P. R. China
| | - Junfei Zhou
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization
- School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing 210023
- P. R. China
| | - Liping Zeng
- Department of Pathogen Biology
- Jiangsu Key Laboratory of Pathogen Biology
- Nanjing Medical University
- Nanjing 211166
- P. R. China
| | - Jingchen Xu
- Department of Pathogen Biology
- Jiangsu Key Laboratory of Pathogen Biology
- Nanjing Medical University
- Nanjing 211166
- P. R. China
| | - Monday M. Onakpa
- Department of Veterinary Pharmacology and Toxicology
- University of Abuja
- Abuja 920001
- Nigeria
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization
- School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing 210023
- P. R. China
| | - Chun-Tao Che
- Department of Pharmaceutical Sciences
- College of Pharmacy
- University of Illinois at Chicago
- Chicago
- USA
| | - Hongkai Bi
- Department of Pathogen Biology
- Jiangsu Key Laboratory of Pathogen Biology
- Nanjing Medical University
- Nanjing 211166
- P. R. China
| | - Ming Zhao
- Jiangsu Collaborative Innovation Centre of Chinese Medicinal Resources Industrialization
- School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing 210023
- P. R. China
| |
Collapse
|
18
|
Henriques PC, Costa LM, Seabra CL, Antunes B, Silva-Carvalho R, Junqueira-Neto S, Maia AF, Oliveira P, Magalhães A, Reis CA, Gartner F, Touati E, Gomes J, Costa P, Martins MCL, Gonçalves IC. Orally administrated chitosan microspheres bind Helicobacter pylori and decrease gastric infection in mice. Acta Biomater 2020; 114:206-220. [PMID: 32622054 DOI: 10.1016/j.actbio.2020.06.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 12/13/2022]
Abstract
Persistent Helicobacter pylori (H. pylori) infection is related to 90% of gastric cancers. With bacterial resistance rising and treatment inefficiency affecting 15% of the patients, alternative treatments urge. Chitosan microspheres (ChMics) have been proposed as an H. pylori-binding system. This work evaluates ChMics biocompatibility, mucopenetration and capacity to treat H. pylori infection in mice after oral administration. ChMics of different size (XL, ∼120 µm and XS, ∼40 µm) and degree of acetylation (6% and 16%) were developed and revealed to be able to adhere both human and mouse-adapted H. pylori strains without cytotoxicity towards human gastric cells. Ex vivo studies showed that smaller (XS) microspheres penetrate further within the gastric foveolae, suggesting their ability to reach deeply adherent bacteria. In vivo assays showed 88% reduction of infection when H. pylori-infected mice (C57BL/6) were treated with more mucoadhesive XL6 and XS6 ChMics. Overall, ChMics clearly demonstrate ability to reduce H. pylori gastric infection in mice, with chitosan degree of acetylation being a dominant factor over microspheres' size on H. pylori removal efficiency. These results evidence the strong potential of this strategy as an antibiotic-free approach to fight H. pylori infection, where microspheres are orally administered, bind H. pylori in the stomach, and remove them through the gastrointestinal tract. STATEMENT OF SIGNIFICANCE: Approximately 90% of gastric cancers are caused by the carcinogenic agent Helicobacter pylori, which infects >50% of the world population. Bacterial resistance, reduced antibiotic bioavailability, and the intricate distribution of bacteria in mucus and within gastric foveolae hamper the success of most strategies to fight H. pylori. We demonstrate that an antibiotic-free therapy based on bare chitosan microspheres that bind and remove H. pylori from stomach can achieve 88% reduction of infection from H. pylori-infected mice. Changing size and mucoadhesive properties, microspheres can reach different areas of gastric mucosa: smaller and less mucoadhesive can penetrate deeper into the foveolae. This promising, simple and inexpensive strategy paves the way for a faster bench-to-bedside transition, therefore holding great potential for clinical application.
Collapse
Affiliation(s)
- Patrícia C Henriques
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Lia M Costa
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Catarina L Seabra
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Bernardo Antunes
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Ricardo Silva-Carvalho
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Susana Junqueira-Neto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - André F Maia
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Pedro Oliveira
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Ana Magalhães
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Celso A Reis
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; Faculdade de Medicina, Universidade do Porto, Al. Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Fátima Gartner
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Eliette Touati
- Unit of Helicobacter Pathogenesis, Department of Microbiology, CNRS UMR2001, Institut Pasteur, 25-28 Rue du Dr. Roux, 75015, Paris, France
| | - Joana Gomes
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, 4200-465 Porto, Portugal
| | - Paulo Costa
- UCIBIO/REQUIMTE, MedTech-Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4150-755 Porto, Portugal
| | - M Cristina L Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Inês C Gonçalves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal.
| |
Collapse
|
19
|
Flavodoxins as Novel Therapeutic Targets against Helicobacter pylori and Other Gastric Pathogens. Int J Mol Sci 2020; 21:ijms21051881. [PMID: 32164177 PMCID: PMC7084853 DOI: 10.3390/ijms21051881] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/04/2020] [Accepted: 03/06/2020] [Indexed: 02/06/2023] Open
Abstract
Flavodoxins are small soluble electron transfer proteins widely present in bacteria and absent in vertebrates. Flavodoxins participate in different metabolic pathways and, in some bacteria, they have been shown to be essential proteins representing promising therapeutic targets to fight bacterial infections. Using purified flavodoxin and chemical libraries, leads can be identified that block flavodoxin function and act as bactericidal molecules, as it has been demonstrated for Helicobacter pylori (Hp), the most prevalent human gastric pathogen. Increasing antimicrobial resistance by this bacterium has led current therapies to lose effectiveness, so alternative treatments are urgently required. Here, we summarize, with a focus on flavodoxin, opportunities for pharmacological intervention offered by the potential protein targets described for this bacterium and provide information on other gastrointestinal pathogens and also on bacteria from the gut microbiota that contain flavodoxin. The process of discovery and development of novel antimicrobials specific for Hp flavodoxin that is being carried out in our group is explained, as it can be extrapolated to the discovery of inhibitors specific for other gastric pathogens. The high specificity for Hp of the antimicrobials developed may be of help to reduce damage to the gut microbiota and to slow down the development of resistant Hp mutants.
Collapse
|
20
|
Abstract
The main challenge in the field of Helicobacter pylori (H. pylori) infection is antibiotic resistance, which influences the efficacy of eradication regimens. Bismuth-containing quadruple therapy has been confirmed as an effective regimen for eradicating H. pylori, especially in strains with antibiotic resistance. High-dose proton-pump inhibitor-amoxicillin dual therapy could decrease the use of unnecessary antibiotics, which is a promising alternative approach. Adjuvant therapy (specific probiotic or vitamin) also showed good results, although more evidence is needed. Novel anti-H. pylori drugs are needed, and the establishment of the H. pylori database is an effective way to acknowledge the real-time information of H. pylori management. This review provides the recent progress of H. pylori treatment, and further studies are needed to address the role of different regimens in improving H. pylori eradication rate, especially in strains with antibiotics resistance.
Collapse
Affiliation(s)
- Yi Hu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | | | | |
Collapse
|
21
|
González A, Casado J, Chueca E, Salillas S, Velázquez-Campoy A, Espinosa Angarica V, Bénejat L, Guignard J, Giese A, Sancho J, Lehours P, Lanas Á. Repurposing Dihydropyridines for Treatment of Helicobacter pylori Infection. Pharmaceutics 2019; 11:pharmaceutics11120681. [PMID: 31847484 PMCID: PMC6969910 DOI: 10.3390/pharmaceutics11120681] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 12/12/2019] [Accepted: 12/13/2019] [Indexed: 12/11/2022] Open
Abstract
Antibiotic resistance is a major cause of the increasing failures in the current eradication therapies against Helicobacter pylori. In this scenario, repurposing drugs could be a valuable strategy to fast-track novel antimicrobial agents. In the present study, we analyzed the inhibitory capability of 1,4-dihydropyridine (DHP) antihypertensive drugs on the essential function of the H. pylori response regulator HsrA and investigated both the in vitro antimicrobial activities and the in vivo efficacy of DHP treatments against H. pylori. Six different commercially available and highly prescribed DHP drugs-namely, Nifedipine, Nicardipine, Nisoldipine, Nimodipine, Nitrendipine, and Lercanidipine-noticeably inhibited the DNA binding activity of HsrA and exhibited potent bactericidal activities against both metronidazole- and clarithromycin-resistant strains of H. pylori, with minimal inhibitory concentration (MIC) values in the range of 4 to 32 mg/L. The dynamics of the decline in the bacterial counts at 2 × MIC appeared to be correlated with the lipophilicity of the drugs, suggesting different translocation efficiencies of DHPs across the bacterial membrane. Oral treatments with 100 mg/kg/day of marketed formulations of Nimodipine or Nitrendipine in combination with omeprazole significantly reduced the H. pylori gastric colonization in mice. The results presented here support a novel therapeutic solution for treatment of antibiotic-resistant H. pylori infections.
Collapse
Affiliation(s)
- Andrés González
- Aragon Institute for Health Research (IIS Aragón), San Juan Bosco 13, 50009 Zaragoza, Spain
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Mariano Esquilor (Edif. I+D), 50018 Zaragoza, Spain
- Correspondence: ; Tel.: +34-976-762807
| | - Javier Casado
- Department of Biochemistry and Molecular & Cellular Biology, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Eduardo Chueca
- Aragon Institute for Health Research (IIS Aragón), San Juan Bosco 13, 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Sandra Salillas
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Mariano Esquilor (Edif. I+D), 50018 Zaragoza, Spain
- Department of Biochemistry and Molecular & Cellular Biology, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Adrián Velázquez-Campoy
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Mariano Esquilor (Edif. I+D), 50018 Zaragoza, Spain
- Department of Biochemistry and Molecular & Cellular Biology, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
- ARAID Foundation, Government of Aragon, Ranillas 1-D, 50018 Zaragoza, Spain
| | - Vladimir Espinosa Angarica
- Cancer Science Institute, National University of Singapore, 14 Medical Drive, #12-01, Singapore 117599, Singapore
| | - Lucie Bénejat
- UMR1053 Bordeaux Research in Translational Oncology, INSERM, Université Bordeaux, BaRITOn, 33000 Bordeaux, France
- French National Reference Center for Campylobacters & Helicobacters, 33000 Bordeaux, France
| | - Jérome Guignard
- UMR1053 Bordeaux Research in Translational Oncology, INSERM, Université Bordeaux, BaRITOn, 33000 Bordeaux, France
| | - Alban Giese
- UMR1053 Bordeaux Research in Translational Oncology, INSERM, Université Bordeaux, BaRITOn, 33000 Bordeaux, France
| | - Javier Sancho
- Aragon Institute for Health Research (IIS Aragón), San Juan Bosco 13, 50009 Zaragoza, Spain
- Institute for Biocomputation and Physics of Complex Systems (BIFI), Mariano Esquilor (Edif. I+D), 50018 Zaragoza, Spain
- Department of Biochemistry and Molecular & Cellular Biology, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Philippe Lehours
- UMR1053 Bordeaux Research in Translational Oncology, INSERM, Université Bordeaux, BaRITOn, 33000 Bordeaux, France
- French National Reference Center for Campylobacters & Helicobacters, 33000 Bordeaux, France
| | - Ángel Lanas
- Aragon Institute for Health Research (IIS Aragón), San Juan Bosco 13, 50009 Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Monforte de Lemos 3-5, 28029 Madrid, Spain
- Digestive Diseases Service, University Clinic Hospital Lozano Blesa; San Juan Bosco 15, 50009 Zaragoza, Spain
- Department of Medicine, Psychiatry and Dermatology, University of Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
| |
Collapse
|
22
|
Wang B, Zhang J, Chen S, Bie M. Helicobacter pylori and gastrointestinal and neurological diseases: Study protocol of an umbrella review of systematic reviews and meta-analyses. Medicine (Baltimore) 2019; 98:e18460. [PMID: 31876728 PMCID: PMC6946528 DOI: 10.1097/md.0000000000018460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 11/18/2019] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Systematic reviews showed that Helicobacter pylori (HP) infection is a major risk for developing gastric cancer and gastric ulcer and that it might be the cause of inflammatory bowel diseases, functional gastrointestinal disorders, and neurological diseases like Alzheimer disease. However, the robustness of the evidence was not tested. We will perform an umbrella review to systematically evaluate current evidence on the correlation between HP infection and gastrointestinal and neurological diseases. METHODS We will search OVID MEDLINE, EMBASE, and the Cochrane library for systematic reviews that evaluate the correlation of HP with gastrointestinal and neurological diseases, from inception to 1 July, 2019. Two reviewers will independently screen titles and abstracts of retrieved articles for eligible studies, and they will extract information for data analysis. We will assess heterogeneity between studies using I statistics and evaluate small-study effect in each systematic review through Egger test. Excess significance bias will be evaluated by compared the expected number of clinical studies with positive findings with the observed number. Quality of each systematic review will be assessed by using AMSTAR2 checklist. ETHICS AND DISSEMINATION This umbrella review is anticipated to be finished in December 2019, and the results will be published in a peer-reviewed journal and disseminated through conference presentation or poster. Because all of the data used in this systematic review and meta-analysis has been published, this review does not require ethical approval.Registration: PROSPERO CRD42019137226.
Collapse
Affiliation(s)
- Baoning Wang
- West China School of Basic Medical Sciences and Forensic Medicine
| | - Jing Zhang
- West China School of Basic Medical Sciences and Forensic Medicine
| | - Sihan Chen
- Jane Lab, Big Data Research Center, University of Electronic Science and Technology of China, Chengdu
| | - Mingjiang Bie
- West China School of Basic Medical Sciences and Forensic Medicine
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, Sichuan
- Editorial Board of Journal of Sichuan University (Medical Science Edition), Chengdu, People's Republic of China
| |
Collapse
|
23
|
Boyanova L, Hadzhiyski P, Kandilarov N, Markovska R, Mitov I. Multidrug resistance in Helicobacter pylori: current state and future directions. Expert Rev Clin Pharmacol 2019; 12:909-915. [PMID: 31424296 DOI: 10.1080/17512433.2019.1654858] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Helicobacter pylori antibiotic resistance has increased worldwide and multidrug resistance (MDR), which seriously hampers eradication success of the frequent chronic infection, has often been reported. Areas covered: H. pylori MDR rates are discussed, mostly from recent articles published since 2015. Present approaches and future directions to counteract the MDR are outlined. Expert opinion: Alarming presence of triple, quadruple and, in some studies, quintuple and sextuple resistance was detected. Primary MDR rates ranged from <10% in most European countries to >40% in Peru. Post-treatment or overall MDR rates were >23-36% in about half of the studies. MDR prevalence has varied both among and within the countries. Factors linked to the MDR are national antibiotic consumption, antibiotic misuse, treatment failures and bacterial factors such as mutations, efflux pumps, and biofilms. Important directions to counteract the MDR increase can be optimization of present and new eradication regimens, wider use of bismuth-containing regimens, assessment of benefit of vonoprazan, new antibiotics such as newer fluoroquinolones and oxazolidinone analogues, adjuvants involving N-acetylcysteine and probiotics, anti-biofilm approaches using anti-biofilm peptides and rhamnolipid and development of vaccines and non-invasive tests for resistance detection. However, more efforts and studies are required. Strain susceptibility testing is increasingly important.
Collapse
Affiliation(s)
- Lyudmila Boyanova
- Department of Medical Microbiology, Medical University of Sofia , Sofia , Bulgaria
| | - Petyo Hadzhiyski
- Specialized Hospital for Active Pediatric Treatment, Medical University of Sofia , Sofia , Bulgaria
| | - Nayden Kandilarov
- Department of General and Hepatobiliary Pancreatic Surgery, Department of Surgery, Medical University , Sofia , Bulgaria
| | - Rumyana Markovska
- Department of Medical Microbiology, Medical University of Sofia , Sofia , Bulgaria
| | - Ivan Mitov
- Department of Medical Microbiology, Medical University of Sofia , Sofia , Bulgaria
| |
Collapse
|