1
|
Ali AH, Hachem M, Ahmmed MK. Docosahexaenoic acid-loaded nanoparticles: A state-of-the-art of preparation methods, characterization, functionality, and therapeutic applications. Heliyon 2024; 10:e30946. [PMID: 38774069 PMCID: PMC11107210 DOI: 10.1016/j.heliyon.2024.e30946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 05/08/2024] [Accepted: 05/08/2024] [Indexed: 05/24/2024] Open
Abstract
Docosahexaenoic acid (DHA, C22:6 n-3), an omega-3 polyunsaturated fatty acid, offers several beneficial effects. DHA helps in reducing depression, autoimmune diseases, rheumatoid arthritis, attention deficit hyperactivity syndrome, and cardiovascular diseases. It can stimulate the development of brain and nerve, alleviate lipids metabolism-related disorders, and enhance vision development. However, DHA susceptibility to chemical oxidation, poor water solubility, and unpleasant order could restrict its applications for nutritional and therapeutic purposes. To avoid these drawbacks and enhance its bioavailability, DHA can be encapsulated using an effective delivery system. Several encapsulation methods are recognized, and DHA-loaded nanoparticles have demonstrated numerous benefits. In clinical studies, positive influences on the development of several diseases have been reported, but some assumptions are conflicting and need more exploration, since DHA has a systemic and not a targeted release at the required level. This might cause the applications of nanoparticles that could allow DHA release at the required level and improve its efficiency, thus resulting in a better controlling of several diseases. In the current review, we focused on researches investigating the formulation and development of DHA-loaded nanoparticles using different delivery systems, including low-density lipoprotein, zinc oxide, silver, zein, and resveratrol-stearate. Silver-DHA nanoparticles presented a typical particle size of 24 nm with an incorporation level of 97.67 %, while the entrapment efficiency of zinc oxide-DHA nanoparticles represented 87.3 %. By using zein/Poly (lactic-co-glycolic acid) stabilized nanoparticles, DHA's encapsulation level reached 84.6 %. We have also highlighted the characteristics, functionality and medical implementation of these nanoparticles in the treatment of inflammations, brain disorders, diabetes as well as hepatocellular carcinoma.
Collapse
Affiliation(s)
- Abdelmoneim H. Ali
- Department of Chemical and Petroleum Engineering, Khalifa University of Science and Technology, Abu Dhabi, 127788, United Arab Emirates
| | - Mayssa Hachem
- Department of Chemistry and Healthcare Engineering Innovation Group, Khalifa University of Sciences and Technology, Abu Dhabi, 127788, United Arab Emirates
| | - Mirja Kaizer Ahmmed
- Department of Fishing and Post-harvest Technology, Chattogram Veterinary and Animal Sciences University, Chattogram, Bangladesh
- Riddet Institute, Massey University, Palmerston North, New Zealand
| |
Collapse
|
2
|
Huang TT, Liu YN, Huang JX, Yan PP, Wang JJ, Cao YX, Cao L. Sodium sulfite-driven Helicobacter pylori eradication: Unraveling oxygen dynamics through multi-omics investigation. Biochem Pharmacol 2024; 222:116055. [PMID: 38354959 DOI: 10.1016/j.bcp.2024.116055] [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: 12/21/2023] [Revised: 01/05/2024] [Accepted: 02/09/2024] [Indexed: 02/16/2024]
Abstract
Due to the emergence and spread of multidrug resistance in Helicobacter pylori (H. pylori), its eradication has become difficult. Sodium sulfite (SS), a widely used food additive for ensuring food safety and storage, has been recognized as an effective nonbactericidal agent for H. pylori eradication. However, the mechanism by which H. pylori adapts and eventually succumbs under low- or no-oxygen conditions remains unknown. In this study, we aimed to evaluate the anti-H. pylori effect of SS and investigated the multiomics mechanism by which SS kills H. pylori. The results demonstrated that SS effectively eradicated H. pylori both in vitro and in vivo. H. pylori responds to the oxygen changes regulated by SS, downregulates the HcpE gene, which is responsible for redox homeostasis in bacteria, decreases the activities of enzymes related to oxidative stress, and disrupts the outer membrane structure, increasing susceptibility to oxidative stress. Furthermore, SS downregulates the content of cytochrome C in the microaerobic respiratory chain, leading to a sharp decrease in ATP synthesis. Consequently, the accumulation of triglycerides (TGs) in bacteria due to oxidative stress supports anaerobic respiration, meeting their energy requirements. The multifaceted death of H. pylori caused by SS does not result in drug resistance. Thus, screening of the redox homeostasis of HcpE as a new target for H. pylori infection treatment could lead to the development of a novel approach for H. pylori eradication therapy.
Collapse
Affiliation(s)
- Ting-Ting Huang
- Department of Pharmacology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Yan-Ni Liu
- Department of Pharmacology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Jin-Xian Huang
- Software Department, East China University of Technology, Nanchang 330032, Jiangxi, China
| | - Ping-Ping Yan
- Department of Pharmacology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China
| | - Ji-Jing Wang
- Department of Medical Biophysics and Biochemistry, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Yong-Xiao Cao
- Department of Pharmacology, School of Basic Medical Science, Xi'an Jiaotong University Health Science Center, Xi'an 710061, Shaanxi, China.
| | - Lei Cao
- Precision Medical Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi, China.
| |
Collapse
|
3
|
Fonseca D, Alves PM, Neto E, Custódio B, Guimarães S, Moura D, Annis F, Martins M, Gomes A, Teixeira C, Gomes P, Pereira RF, Freitas P, Parreira P, Martins MCL. One-Pot Microfluidics to Engineer Chitosan Nanoparticles Conjugated with Antimicrobial Peptides Using "Photoclick" Chemistry: Validation Using the Gastric Bacterium Helicobacter pylori. ACS APPLIED MATERIALS & INTERFACES 2024; 16:14533-14547. [PMID: 38482690 PMCID: PMC10982938 DOI: 10.1021/acsami.3c18772] [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: 12/14/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024]
Abstract
Surface bioconjugation of antimicrobial peptides (AMP) onto nanoparticles (AMP-NP) is a complex, multistep, and time-consuming task. Herein, a microfluidic system for the one-pot production of AMP-NP was developed. Norbornene-modified chitosan was used for NP production (NorChit-NP), and thiolated-AMP was grafted on their surface via thiol-norbornene "photoclick" chemistry over exposure of two parallel UV LEDs. The MSI-78A was the AMP selected due to its high activity against a high priority (level 2) antibiotic-resistant gastric pathogen: Helicobacter pylori (H. pylori). AMP-NP (113 ± 43 nm; zeta potential 14.3 ± 7 mV) were stable in gastric settings without a cross-linker (up to 5 days in pH 1.2) and bactericidal against two highly pathogenic H. pylori strains (1011 NP/mL with 96 μg/mL MSI-78A). Eradication was faster for H. pylori 26695 (30 min) than for H. pylori J99 (24 h), which was explained by the lower minimum bactericidal concentration of soluble MSI-78A for H. pylori 26695 (32 μg/mL) than for H. pylori J99 (128 μg/mL). AMP-NP was bactericidal by inducing H. pylori cell membrane alterations, intracellular reorganization, generation of extracellular vesicles, and leakage of cytoplasmic contents (transmission electron microscopy). Moreover, NP were not cytotoxic against two gastric cell lines (AGS and MKN74, ATCC) at bactericidal concentrations. Overall, the designed microfluidic setup is a greener, simpler, and faster approach than the conventional methods to obtain AMP-NP. This technology can be further explored for the bioconjugation of other thiolated-compounds.
Collapse
Affiliation(s)
- Diana
R. Fonseca
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto
Nacional de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- Faculdade
de Engenharia, Departamento de Engenharia Metalúrgica e de
Materiais, Universidade do Porto, R. Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Pedro M. Alves
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto
Nacional de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- Faculdade
de Engenharia, Departamento de Engenharia Metalúrgica e de
Materiais, Universidade do Porto, R. Dr. Roberto Frias, 4200-465 Porto, Portugal
- LAQV-REQUIMTE,
Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 685, 4169-007 Porto, Portugal
| | - Estrela Neto
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto
Nacional de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Beatriz Custódio
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto
Nacional de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS−Instituto
de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Sofia Guimarães
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto
Nacional de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Duarte Moura
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto
Nacional de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- Faculdade
de Engenharia, Departamento de Engenharia Metalúrgica e de
Materiais, Universidade do Porto, R. Dr. Roberto Frias, 4200-465 Porto, Portugal
| | - Francesca Annis
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto
Nacional de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
| | - Marco Martins
- INL, International
Iberian Nanotechnology Laboratory, Av. Mte. José Veiga s/n, 4715-330 Braga, Portugal
| | - Ana Gomes
- LAQV-REQUIMTE,
Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 685, 4169-007 Porto, Portugal
| | - Cátia Teixeira
- LAQV-REQUIMTE,
Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 685, 4169-007 Porto, Portugal
| | - Paula Gomes
- LAQV-REQUIMTE,
Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 685, 4169-007 Porto, Portugal
| | - Rúben F. Pereira
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto
Nacional de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS−Instituto
de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| | - Paulo Freitas
- INL, International
Iberian Nanotechnology Laboratory, Av. Mte. José Veiga s/n, 4715-330 Braga, Portugal
- INESC-MN,
INESC Microsystems and Nanotechnologies, Rua Alves Redol 9, 1000-029 Lisboa, Portugal
| | - Paula Parreira
- i3S
− Instituto de Investigação e Inovação
em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
- Instituto
Nacional de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 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
- Instituto
Nacional de Engenharia Biomédica, Universidade do Porto, R. Alfredo Allen 208, 4200-135 Porto, Portugal
- ICBAS−Instituto
de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal
| |
Collapse
|
4
|
Khoshnood S, Negahdari B, Kaviar VH, Sadeghifard N, Abdullah MA, El-Shazly M, Haddadi MH. Amoxicillin-docosahexaenoic acid encapsulated chitosan-alginate nanoparticles as a delivery system with enhanced biocidal activities against Helicobacter pylori and improved ulcer healing. Front Microbiol 2023; 14:1083330. [PMID: 36846798 PMCID: PMC9948253 DOI: 10.3389/fmicb.2023.1083330] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
Encapsulation of amoxicillin (AMX) for drug delivery against Helicobacter pylori infection and aspirin-induced ulcers in rat's stomachs was performed using docosahexaenoic acid (DHA)-loaded chitosan/alginate (CA) nanoparticles (NPs) developed by ionotropic gelation method. The physicochemical analyses of the composite NPs were performed by scanning electron microscopy, Fourier transform infrared spectroscopy, zeta potential, X-ray diffraction, and atomic force microscopy. The encapsulation efficiency of AMX was increased to 76% by incorporating DHA, which resulted in a reduction in the particle size. The formed CA-DHA-AMX NPs effectively adhered to the bacteria and rat gastric mucosa. Their antibacterial properties were more potent than those of the single AMX and CA-DHA NPs as demonstrated by the in vivo assay. The composite NPs attained higher mucoadhesive potential during food intake than during fasting (p = 0.029). At 10 and 20 mg/kg AMX, the CA-AMX-DHA showed more potent activities against H. pylori than the CA-AMX, CA-DHA, and single AMX. The in vivo study showed that the effective dose of AMX was lower when DHA was included, indicating better drug delivery and stability of the encapsulated AMX. Both mucosal thickening and ulcer index were significantly higher in the groups receiving CA-DHA-AMX than in the groups receiving CA-AMX and single AMX. The presence of DHA declines the pro-inflammatory cytokines including IL-1β, IL-6, and IL-17A. The synergistic effects of AMX and the CA-DHA formulation increased the biocidal activities against H. pylori infection and improved ulcer healing properties.
Collapse
Affiliation(s)
- Saeed Khoshnood
- Clinical Microbiology Research Centre, Ilam University of Medical Sciences, Ilam, Iran
| | - Babak Negahdari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahab Hassan Kaviar
- Clinical Microbiology Research Centre, Ilam University of Medical Sciences, Ilam, Iran
| | - Nourkhoda Sadeghifard
- Clinical Microbiology Research Centre, Ilam University of Medical Sciences, Ilam, Iran
| | - Mohd Azmuddin Abdullah
- Department of Toxicology, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Mohamed El-Shazly
- Department of Pharmacognosy, Faculty of Pharmacy, Ain-Shams University, Cairo, Egypt
| | - Mohammad Hossein Haddadi
- Clinical Microbiology Research Centre, Ilam University of Medical Sciences, Ilam, Iran,*Correspondence: Mohammad Hossein Haddadi,✉ ;✉
| |
Collapse
|
5
|
Helicobacter pylori biofilms are disrupted by nanostructured lipid carriers: A path to eradication? J Control Release 2022; 348:489-498. [PMID: 35654169 DOI: 10.1016/j.jconrel.2022.05.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 11/20/2022]
Abstract
Bacterial biofilms account for 80% of all chronic infections, with cells being up to 1000 times more resistant to antibiotics than their planktonic counterparts. The recently discovered ability of Helicobacter pylori to form biofilms once again highlights why this bacterium is one of the most successful human pathogens. The current treatments failure rate reaches 40% of cases, emphasizing that new therapeutic options are a pressing need. Nanostructured lipid carriers (NLC), with and without docosahexaenoic acid (DHA), were very effective against H. pylori planktonic cells but their effect on H. pylori biofilms was unknown. Here, DHA-loaded NLC (DHA-NLC) and NLC without any drug (blank NLC) were tested on an optimized H. pylori in vitro floating mature biofilm model. DHA-NLC and blank NLC reduced the total biofilm biomass and had a bactericidal effect against both biofilm and planktonic bacteria in all the concentrations tested (0.125-2 mg/mL). DHA-NLC achieved biofilm biomass reduction in a concentration ~ 8 times lower than blank NLC (0.125 vs 1 mg/mL, respectively). Both NLC were bactericidal at the lowest concentration tested (0.125 mg/mL) although with different efficiency, i.e. a decrease of ∼6 log10 for DHA-NLC and ∼5 log10 for blank NLC. In addition, the equivalent amount of free DHA (3.1 μM) only reduced bacterial viability in ∼2 log10, demonstrating the synergistic effect of DHA and NLC in the treatment of H. pylori biofilms. Nevertheless, although viable bacteria were not detected by colony forming unit (CFU) counting after treatment with both NLC, confocal microscopy imaging highlighted that some H. pylori cells remained alive. In addition, scanning electron microscopy (SEM) analysis confirmed an increase in bacteria with a coccoid morphology after treatment, suggesting a transition to a viable but non-culturable (VBNC) state. Altogether, it is herein established that NLC, even without any drug, are promising for the management of H. pylori bacteria organized in biofilms, opening new perspectives for the eradication of this gastric pathogen.
Collapse
|
6
|
White B, Sterrett JD, Grigoryan Z, Lally L, Heinze JD, Alikhan H, Lowry CA, Perez LJ, DeSipio J, Phadtare S. Characterization of gut microbiome and metabolome in Helicobacter pylori patients in an underprivileged community in the United States. World J Gastroenterol 2021; 27:5575-5594. [PMID: 34588753 PMCID: PMC8433610 DOI: 10.3748/wjg.v27.i33.5575] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/02/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori), a bacterium that infects approximately half of the world's population, is associated with various gastrointestinal diseases, including peptic ulcers, non-ulcer dyspepsia, gastric adenocarcinoma, and gastric lymphoma. As the burden of antibiotic resistance increases, the need for new adjunct therapies designed to facilitate H. pylori eradication and reduce negative distal outcomes associated with infection has become more pressing. Characterization of the interactions between H. pylori, the fecal microbiome, and fecal fatty acid metabolism, as well as the mechanisms underlying these interactions, may offer new therapeutic approaches. AIM To characterize the gut microbiome and metabolome in H. pylori patients in a socioeconomically challenged and underprivileged inner-city community. METHODS Stool samples from 19 H. pylori patients and 16 control subjects were analyzed. 16S rRNA gene sequencing was performed on normalized pooled amplicons using the Illumina MiSeq System using a MiSeq reagent kit v2. Alpha and beta diversity analyses were performed in QIIME 2. Non-targeted fatty acid analysis of the samples was carried out using gas chromatography-mass spectrometry, which measures the total content of 30 fatty acids in stool after conversion into their corresponding fatty acid methyl esters. Multi-dimensional scaling (MDS) was performed on Bray-Curtis distance matrices created from both the metabolomics and microbiome datasets and a Procrustes test was performed on the metabolomics and microbiome MDS coordinates. RESULTS Fecal microbiome analysis showed that alpha diversity was lowest in H. pylori patients over 40 years of age compared to control subjects of similar age group. Beta diversity analysis of the samples revealed significant differences in microbial community structure between H. pylori patients and control subjects across all ages. Thirty-eight and six taxa had lower and higher relative abundance in H. pylori patients, respectively. Taxa that were enriched in H. pylori patients included Atopobium, Gemellaceae, Micrococcaceae, Gemellales and Rothia (R. mucilaginosa). Notably, relative abundance of the phylum Verrucomicrobia was decreased in H. pylori patients compared to control subjects. Procrustes analysis showed a significant relationship between the microbiome and metabolome datasets. Stool samples from H. pylori patients showed increases in several fatty acids including the polyunsaturated fatty acids (PUFAs) 22:4n6, 22:5n3, 20:3n6 and 22:2n6, while decreases were noted in other fatty acids including the PUFA 18:3n6. The pattern of changes in fatty acid concentration correlated to the Bacteroidetes:Firmicutes ratio determined by 16S rRNA gene analysis. CONCLUSION This exploratory study demonstrates H. pylori-associated changes to the fecal microbiome and fecal fatty acid metabolism. Such changes may have implications for improving eradication rates and minimizing associated negative distal outcomes.
Collapse
Affiliation(s)
- Brian White
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| | - John D Sterrett
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, United States
| | - Zoya Grigoryan
- Department of Internal Medicine, Lenox Hill Hospital, NYC, NY 10075, United States
| | - Lauren Lally
- Department of Internal Medicine, Thomas Jefferson University Hospital, Philadelphia, PA 19107, United States
| | - Jared D Heinze
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, United States
| | - Hyder Alikhan
- Department of Biological Sciences, Rowan University, Glassboro, NJ 08028, United States
| | - Christopher A Lowry
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, United States
| | - Lark J Perez
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ 08028, United States
| | - Joshua DeSipio
- Department of Gastroenterology, Cooper University Hospital, Camden, NJ 08103, United States
| | - Sangita Phadtare
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, United States
| |
Collapse
|
7
|
Coraça-Huber DC, Steixner S, Wurm A, Nogler M. Antibacterial and Anti-Biofilm Activity of Omega-3 Polyunsaturated Fatty Acids against Periprosthetic Joint Infections-Isolated Multi-Drug Resistant Strains. Biomedicines 2021; 9:biomedicines9040334. [PMID: 33810261 PMCID: PMC8065983 DOI: 10.3390/biomedicines9040334] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 12/19/2022] Open
Abstract
Background: Implantable medical devices, such as prosthetics, catheters, and several other devices, have revolutionized medicine, but they increase the infection risk. In previous decades, commercially available antibiotics lost their activity against coagulase-negative Staphylococci (CoNS) and several other microorganisms. Docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) are the two major omega-3 polyunsaturated fatty acids (ω-3 PUFAs) with antimicrobial properties. Materials and Methods: In this study, we tested the EPA and the DHA for its antibacterial and anti-biofilm activity in vitro against Staphylococcus epidermidis, Staphylococcus aureus, and different CoNS as reference strains and isolated from patients undergoing orthopedic treatment for implant infections. The tests were carried out with the strains in planktonic and biofilm form. Cytotoxicity assay was carried out with EPA and DHA using human gingival fibroblasts HGF-1. Results: The highest concentration of EPA and DHA promoted the complete killing of S. epidermidis 1457 and S. aureus ATCC 25923 in planktonic form. The fatty acids showed low activity against P. aeruginosa. EPA and DHA completely killed or significantly reduced the count of planktonic bacteria of the patient isolated strains. When incubated with media enriched with EPA and DHA, the biofilm formation was significantly reduced on S. epidermidis 1457 and not present on S. aureus ATCC 25923. The reduction or complete killing were also observed with the clinical isolates. The pre-formed biofilms showed reduction of the cell counting after treatment with EPA and DHA. Conclusion: In this study, the ω-3 PUFAs EPA and DHA showed antimicrobial and anti-biofilm activity in vitro against S. aureus, S. epidermidis, and P. aeruginosa, as well as against multi-drug resistant S. aureus and CoNS strains isolated from patients undergoing periprosthetic joint infections (PJI) treatment. Higher concentrations of the fatty acids showed killing activity on planktonic cells and inhibitory activity of biofilm formation. Although both substances showed antimicrobial activity, EPA showed better results in comparison with DHA. In addition, when applied on human gingival fibroblasts in vitro, EPA and DHA showed a possible protective effect on cells cultured in medium enriched with ethanol. Further studies are required to confirm the antimicrobial activity of EPA and DHA against multi-drug resistant strains and pan-drug resistant strains.
Collapse
Affiliation(s)
- Débora C. Coraça-Huber
- Research Laboratory for Biofilms and Implant Associated Infections (BIOFILM LAB), Experimental Orthopedics, University Hospital for Orthopedics and Traumatology, Medical University of Innsbruck, Peter-Mayr-Strasse 4b, Room 204, 6020 Innsbruck, Austria; (S.S.); (M.N.)
- Correspondence: ; Tel.: +43-512-9003-71697; Fax: +43-512-9003-73691
| | - Stephan Steixner
- Research Laboratory for Biofilms and Implant Associated Infections (BIOFILM LAB), Experimental Orthopedics, University Hospital for Orthopedics and Traumatology, Medical University of Innsbruck, Peter-Mayr-Strasse 4b, Room 204, 6020 Innsbruck, Austria; (S.S.); (M.N.)
| | - Alexander Wurm
- University Hospital for Orthopedics and Traumatology, Medical University of Innsbruck, Anichstraße 35, 6020 Innsbruck, Austria;
| | - Michael Nogler
- Research Laboratory for Biofilms and Implant Associated Infections (BIOFILM LAB), Experimental Orthopedics, University Hospital for Orthopedics and Traumatology, Medical University of Innsbruck, Peter-Mayr-Strasse 4b, Room 204, 6020 Innsbruck, Austria; (S.S.); (M.N.)
| |
Collapse
|
8
|
Zare Javid A, Maghsoumi-Norouzabad L, Bazyar H, Aghamohammadi V, Alavinejad P. Effects of Concurrent Omega-3 and Cranberry Juice Consumption Along with Standard Antibiotic Therapy on the Eradication of Helicobacter p ylori, Gastrointestinal Symptoms, Some Serum Inflammatory and Oxidative Stress Markers in Adults with Helicobacter p ylori Infection: A Study Protocol for a Randomized Controlled Trial. Infect Drug Resist 2020; 13:3179-3185. [PMID: 32982332 PMCID: PMC7508030 DOI: 10.2147/idr.s270057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/03/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND It seems alternative treatments such as antioxidant intervention and anti-inflammatory intervention adjuvant to antibiotic regimens may enhance cancer prevention approaches and decrease adverse side effects related to therapeutic antibiotic regimens. So, we will evaluate the effects of concurrent omega-3 and cranberry juice supplementation along with standard antibiotic therapy on the eradication of Helicobacter pylori, gastrointestinal symptoms, some serum inflammatory and oxidative stress markers in adults with HP infection. METHODS We will conduct a 4-week double-blinded randomized clinical trial. The subjects will be randomly stratified according to sex and BMI using a permuted block randomization procedure by Random Allocation Software (RAS). They will be assigned to one of the four study groups: (1) cranberry juice fortified with omega-3 Intervention (n=23), (2) cranberry juice intervention group (n=23), (3) placebo beverage fortified with omega-3 intervention group (n=23), and (4) placebo beverage intervention (control group) (n=23). All statistical analyses will be performed using IBM SPSS Statistics software. DISCUSSION A combination of alternative therapies may have a synergistic effect compared to a single approach. It could potentially be more effective in promoting the efficacy of standard antibiotic therapy in eradicating HP infection. TRIAL REGISTRATION Iranian Registry of Clinical Trials (IRCT20151128025274N3, www.irct.ir/trial/28997). TRIAL STATUS This study is in the early stages of sampling.
Collapse
Affiliation(s)
- Ahmad Zare Javid
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Leila Maghsoumi-Norouzabad
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Nutrition, School of Allied Medical Sciences, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hadi Bazyar
- Nutrition and Metabolic Diseases Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Pejman Alavinejad
- Alimentary Tract Research Center, Imam Khomeini Hospital Clinical Research Development Unit, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
9
|
Ribeiro-Vidal H, Sánchez MC, Alonso-Español A, Figuero E, Ciudad MJ, Collado L, Herrera D, Sanz M. Antimicrobial Activity of EPA and DHA against Oral Pathogenic Bacteria Using an In Vitro Multi-Species Subgingival Biofilm Model. Nutrients 2020; 12:nu12092812. [PMID: 32937742 PMCID: PMC7551721 DOI: 10.3390/nu12092812] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/09/2020] [Accepted: 09/10/2020] [Indexed: 11/16/2022] Open
Abstract
In search for natural products with antimicrobial properties for use in the prevention and treatment of periodontitis, the purpose of this investigation was to evaluate the antimicrobial activity of two omega-3 fatty acids, docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), using an in vitro multi-species subgingival biofilm model including Streptococcus oralis, Actinomyces naeslundii, Veillonella parvula, Fusobacterium nucleatum, Porphyromonas gingivalis, and Aggregatibacter actinomycetemcomitans. The antimicrobial activities of EPA and DHA extracts (100 µM) and the respective controls were assessed on 72 h biofilms by their submersion onto discs for 60 s. Antimicrobial activity was evaluated by quantitative polymerase chain reaction (qPCR), confocal laser scanning microscopy (CLSM) and scanning electron microscopy (SEM). ANOVA with Bonferroni correction was used to evaluate the antimicrobial activity of each of the fatty acids. Both DHA and EPA significantly reduced (p < 0.001 in all cases) the bacterial strains used in this biofilm model. The results with CLSM were consistent with those reported with qPCR. Structural damage was evidenced by SEM in some of the observed bacteria. It was concluded that both DHA and EPA have significant antimicrobial activity against the six bacterial species included in this biofilm model.
Collapse
Affiliation(s)
- Honorato Ribeiro-Vidal
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, University Complutense of Madrid, 28040 Madrid, Spain; (H.R.-V.); (M.C.S.); (A.A.-E.); (E.F.); (D.H.)
| | - María Carmen Sánchez
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, University Complutense of Madrid, 28040 Madrid, Spain; (H.R.-V.); (M.C.S.); (A.A.-E.); (E.F.); (D.H.)
- Medicine Department, Faculty of Medicine, University Complutense of Madrid, 28040 Madrid, Spain; (M.J.C.); (L.C.)
| | - Andrea Alonso-Español
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, University Complutense of Madrid, 28040 Madrid, Spain; (H.R.-V.); (M.C.S.); (A.A.-E.); (E.F.); (D.H.)
| | - Elena Figuero
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, University Complutense of Madrid, 28040 Madrid, Spain; (H.R.-V.); (M.C.S.); (A.A.-E.); (E.F.); (D.H.)
| | - Maria José Ciudad
- Medicine Department, Faculty of Medicine, University Complutense of Madrid, 28040 Madrid, Spain; (M.J.C.); (L.C.)
| | - Luís Collado
- Medicine Department, Faculty of Medicine, University Complutense of Madrid, 28040 Madrid, Spain; (M.J.C.); (L.C.)
| | - David Herrera
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, University Complutense of Madrid, 28040 Madrid, Spain; (H.R.-V.); (M.C.S.); (A.A.-E.); (E.F.); (D.H.)
| | - Mariano Sanz
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, University Complutense of Madrid, 28040 Madrid, Spain; (H.R.-V.); (M.C.S.); (A.A.-E.); (E.F.); (D.H.)
- Correspondence: ; Tel.: +34-913-942-021
| |
Collapse
|
10
|
Benedetti I, Hoyos J, Barrios L. Cyclooxigenase-2 and osteopontin in gastric pre-neoplastic lesions in relation to H-pylori infection and grade of inflammation. REVISTA ESPAÑOLA DE PATOLOGÍA : PUBLICACIÓN OFICIAL DE LA SOCIEDAD ESPAÑOLA DE ANATOMÍA PATOLÓGICA Y DE LA SOCIEDAD ESPAÑOLA DE CITOLOGÍA 2020; 53:79-87. [PMID: 32199598 DOI: 10.1016/j.patol.2019.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/20/2019] [Accepted: 06/13/2019] [Indexed: 02/07/2023]
Abstract
The association between Helicobacter-pylori-induced inflammation and gastric adenocarcinoma is well documented and it has been suggested that the pro-mitotic and apoptotic effect of Cyclooxygenase-2 and Osteopontin on the epithelial cells of the gastric mucosa may have a role in carcinogenesis of the gastric mucosa. The aim of this study was to determine the expression of Cyclooxygenase-2 and Osteopontin in normal gastric mucosa, mucosa with gastritis and gastric mucosa with intestinal metaplasia, in relation to Helicobacter-pylori infection and grade of inflammation. Immunohistochemistry was performed on 108 gastric biopsies in order to detect Cyclooxygenase-2 and Osteopontin expression. The intensity and percentage of staining were evaluated using the H-Score, and its association with grade of inflammation, Helicobacter pylori infection and intestinal metaplasia was determined. Expression of Cyclooxygenase-2 and Osteopontin was higher in gastric biopsies (values shown respectively) with Helicobacter-pylori infection (179.9/142.3), intestinal metaplasia (208.8/179.3) or higher grades of inflammation (190/135.7) in comparison to normal gastric mucosa (100.7/80) or mild grade of inflammation (128.4/128.4), (p<0.05).There is an overexpression of Cyclooxygenase-2 and Osteopontin in gastric mucosa with H. pylori infection, intestinal metaplasia and high grades of inflammation, suggesting a constant up-regulation of protein expression in response to the inflammatory process generated by a Helicobacter-pylori infection, leading to the development of intestinal metaplasia.
Collapse
Affiliation(s)
- Inés Benedetti
- Grupo de investigación Histopatología, Facultad de Medicina, Universidad de Cartagena, Carrera 50 # 24-120, código postal 130014 Cartagena, Colombia.
| | - Julián Hoyos
- Grupo de investigación Histopatología, Facultad de Medicina, Universidad de Cartagena, Carrera 50 # 24-120, código postal 130014 Cartagena, Colombia
| | - Lía Barrios
- Grupo de investigación Histopatología, Facultad de Medicina, Universidad de Cartagena, Carrera 50 # 24-120, código postal 130014 Cartagena, Colombia
| |
Collapse
|
11
|
Yoon BK, Jackman JA, Valle-González ER, Cho NJ. Antibacterial Free Fatty Acids and Monoglycerides: Biological Activities, Experimental Testing, and Therapeutic Applications. Int J Mol Sci 2018. [PMID: 29642500 DOI: 10.3390/ijms19041114.pmid:29642500;pmcid:pmc5979495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
Antimicrobial lipids such as fatty acids and monoglycerides are promising antibacterial agents that destabilize bacterial cell membranes, causing a wide range of direct and indirect inhibitory effects. The goal of this review is to introduce the latest experimental approaches for characterizing how antimicrobial lipids destabilize phospholipid membranes within the broader scope of introducing current knowledge about the biological activities of antimicrobial lipids, testing strategies, and applications for treating bacterial infections. To this end, a general background on antimicrobial lipids, including structural classification, is provided along with a detailed description of their targeting spectrum and currently understood antibacterial mechanisms. Building on this knowledge, different experimental approaches to characterize antimicrobial lipids are presented, including cell-based biological and model membrane-based biophysical measurement techniques. Particular emphasis is placed on drawing out how biological and biophysical approaches complement one another and can yield mechanistic insights into how the physicochemical properties of antimicrobial lipids influence molecular self-assembly and concentration-dependent interactions with model phospholipid and bacterial cell membranes. Examples of possible therapeutic applications are briefly introduced to highlight the potential significance of antimicrobial lipids for human health and medicine, and to motivate the importance of employing orthogonal measurement strategies to characterize the activity profile of antimicrobial lipids.
Collapse
Affiliation(s)
- Bo Kyeong Yoon
- School of Materials Science and Engineering and Centre for Biomimetic Sensor Science, Nanyang Technological University, 50 Nanyang Drive, Singapore 637553, Singapore.
| | - Joshua A Jackman
- School of Materials Science and Engineering and Centre for Biomimetic Sensor Science, Nanyang Technological University, 50 Nanyang Drive, Singapore 637553, Singapore.
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Elba R Valle-González
- School of Materials Science and Engineering and Centre for Biomimetic Sensor Science, Nanyang Technological University, 50 Nanyang Drive, Singapore 637553, Singapore.
| | - Nam-Joon Cho
- School of Materials Science and Engineering and Centre for Biomimetic Sensor Science, Nanyang Technological University, 50 Nanyang Drive, Singapore 637553, Singapore.
| |
Collapse
|
12
|
Yoon BK, Jackman JA, Valle-González ER, Cho NJ. Antibacterial Free Fatty Acids and Monoglycerides: Biological Activities, Experimental Testing, and Therapeutic Applications. Int J Mol Sci 2018; 19:E1114. [PMID: 29642500 PMCID: PMC5979495 DOI: 10.3390/ijms19041114] [Citation(s) in RCA: 314] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 04/05/2018] [Accepted: 04/05/2018] [Indexed: 02/06/2023] Open
Abstract
Antimicrobial lipids such as fatty acids and monoglycerides are promising antibacterial agents that destabilize bacterial cell membranes, causing a wide range of direct and indirect inhibitory effects. The goal of this review is to introduce the latest experimental approaches for characterizing how antimicrobial lipids destabilize phospholipid membranes within the broader scope of introducing current knowledge about the biological activities of antimicrobial lipids, testing strategies, and applications for treating bacterial infections. To this end, a general background on antimicrobial lipids, including structural classification, is provided along with a detailed description of their targeting spectrum and currently understood antibacterial mechanisms. Building on this knowledge, different experimental approaches to characterize antimicrobial lipids are presented, including cell-based biological and model membrane-based biophysical measurement techniques. Particular emphasis is placed on drawing out how biological and biophysical approaches complement one another and can yield mechanistic insights into how the physicochemical properties of antimicrobial lipids influence molecular self-assembly and concentration-dependent interactions with model phospholipid and bacterial cell membranes. Examples of possible therapeutic applications are briefly introduced to highlight the potential significance of antimicrobial lipids for human health and medicine, and to motivate the importance of employing orthogonal measurement strategies to characterize the activity profile of antimicrobial lipids.
Collapse
Affiliation(s)
- Bo Kyeong Yoon
- School of Materials Science and Engineering and Centre for Biomimetic Sensor Science, Nanyang Technological University, 50 Nanyang Drive, Singapore 637553, Singapore.
| | - Joshua A Jackman
- School of Materials Science and Engineering and Centre for Biomimetic Sensor Science, Nanyang Technological University, 50 Nanyang Drive, Singapore 637553, Singapore.
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Elba R Valle-González
- School of Materials Science and Engineering and Centre for Biomimetic Sensor Science, Nanyang Technological University, 50 Nanyang Drive, Singapore 637553, Singapore.
| | - Nam-Joon Cho
- School of Materials Science and Engineering and Centre for Biomimetic Sensor Science, Nanyang Technological University, 50 Nanyang Drive, Singapore 637553, Singapore.
| |
Collapse
|
13
|
Sun M, Dong J, Xia Y, Shu R. Antibacterial activities of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) against planktonic and biofilm growing Streptococcus mutans. Microb Pathog 2017; 107:212-218. [PMID: 28373143 DOI: 10.1016/j.micpath.2017.03.040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 03/29/2017] [Accepted: 03/30/2017] [Indexed: 10/19/2022]
Abstract
The aim of this study was to evaluate the potential antibacterial activities of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) against planktonic and biofilm modes of Streptococcus mutans (S. mutans). The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) were determined. The effects on planktonic growth and biofilm metabolic activity were evaluated by growth curve determination and MTT assay, respectively. Then, colony forming unit (CFU) counting, scanning electron microscopy (SEM) and real-time PCR were performed to further investigate the actions of DHA and EPA on exponential phase-S. mutans. Confocal laser scanning microscopy (CLSM) was used to detect the influences on mature biofilms. The MICs of DHA and EPA against S. mutans were 100 μM and 50 μM, respectively; the MBC of both compounds was 100 μM. In the presence of 12.5 μM-100 μM DHA or EPA, the planktonic growth and biofilm metabolic activity were reduced in varying degrees. For exponential-phase S. mutans, the viable counts, the bacterial membranes and the biofilm-associated gene expression were damaged by 100 μM DHA or EPA treatment. For 1-day-old biofilms, the thickness was decreased and the proportion of membrane-damaged bacteria was increased in the presence of 100 μM DHA or EPA. These results indicated that, DHA and EPA possessed antibacterial activities against planktonic and biofilm growing S. mutans.
Collapse
Affiliation(s)
- Mengjun Sun
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Jiachen Dong
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Yiru Xia
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Rong Shu
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China.
| |
Collapse
|
14
|
Seabra CL, Nunes C, Gomez-Lazaro M, Correia M, Machado JC, Gonçalves IC, Reis CA, Reis S, Martins MCL. Docosahexaenoic acid loaded lipid nanoparticles with bactericidal activity against Helicobacter pylori. Int J Pharm 2017; 519:128-137. [DOI: 10.1016/j.ijpharm.2017.01.014] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/04/2017] [Accepted: 01/06/2017] [Indexed: 02/06/2023]
|
15
|
Pan CY, Liu YH, Gong HY, Chen JY. Transcriptome analysis of the effect of polyunsaturated fatty acids against Vibrio vulnificus infection in Oreochromis niloticus. FISH & SHELLFISH IMMUNOLOGY 2017; 62:153-163. [PMID: 28108339 DOI: 10.1016/j.fsi.2017.01.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Revised: 01/11/2017] [Accepted: 01/13/2017] [Indexed: 06/06/2023]
Abstract
Vibrio vulnificus infection causes severe economic losses in Oreochromis niloticus aquaculture by inducing pro-inflammatory cytokines, that lead to inflammation and mortality. Omega-3 fatty acids, such as Docosahexaenoic acid (DHA) and Eicosapentaenoic acid (EPA), have been reported for their anti-inflammatory and antibacterial abilities in murine and zebrafish models. However, the anti-inflammatory and antibacterial functions of DHA and EPA in commercial aquaculture organisms such as Oreochromis niloticus remain unknown. The present study demonstrates antibacterial function and transcriptional modulation of inflammation-associated genes by DHA and EPA in Vibrio vulnificus infection in Oreochromis niloticus fish models. The administration of EPA or DHA improved the Oreochromis niloticus survival rate against Vibrio vulnificus infection. The induction of proinflammatory cytokines, Interleukin (IL)-1β, IL-6, Tumor necrosis factor (TNF)-α, and Toll-like receptor (TLR)-2 by Vibrio vulnificus was suppressed in fish that were administered DHA. Bacterial membrane disruption and the killing of Vibrio vulnificus by EPA and DHA was observed using SEM, TEM, and cytoplasm leakage studies. In silico analysis of the transcription profile in Ingenuity Pathway Analysis software showed that DHA may enhance anti-Vibrio vulnificus activity in Oreochromis niloticus via the activation of peroxisome proliferator-activated receptor α (PPARα) to inhibit nuclear factor kappa B and suppress hepatocyte nuclear factor 4 α (HNF4α). In summary, the results of the present study demonstrated that DHA and EPA reduce the severity of Vibrio vulnificus infection and increase the survival rate of Oreochromis niloticus.
Collapse
Affiliation(s)
- Chieh-Yu Pan
- Department and Graduate Institute of Aquaculture, National Kaohsiung Marine University, Kaohsiung 811, Taiwan
| | - Yu-Hao Liu
- Department and Graduate Institute of Aquaculture, National Kaohsiung Marine University, Kaohsiung 811, Taiwan
| | - Hong-Yi Gong
- Department of Aquaculture, National Taiwan Ocean University, Keelung 20224, Taiwan
| | - Jyh-Yih Chen
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, 23-10 Dahuen Road, Jiaushi, Ilan 262, Taiwan.
| |
Collapse
|
16
|
Modulation of host defence against bacterial and viral infections by omega-3 polyunsaturated fatty acids. J Infect 2016; 73:523-535. [DOI: 10.1016/j.jinf.2016.10.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 10/07/2016] [Accepted: 10/07/2016] [Indexed: 12/20/2022]
|
17
|
Sun M, Zhou Z, Dong J, Zhang J, Xia Y, Shu R. Antibacterial and antibiofilm activities of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) against periodontopathic bacteria. Microb Pathog 2016; 99:196-203. [PMID: 27565090 DOI: 10.1016/j.micpath.2016.08.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 08/21/2016] [Accepted: 08/22/2016] [Indexed: 01/02/2023]
Abstract
Docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) are two major omega-3 polyunsaturated fatty acids (n-3 PUFAs) with antimicrobial properties. In this study, we evaluated the potential antibacterial and antibiofilm activities of DHA and EPA against two periodontal pathogens, Porphyromonas gingivalis (P. gingivalis) and Fusobacterium nucleatum (F. nucleatum). MTT assay showed that DHA and EPA still exhibited no cytotoxicity to human oral tissue cells when the concentration came to 100 μM and 200 μM, respectively. Against P. gingivalis, DHA and EPA showed the same minimum inhibitory concentration (MIC) of 12.5 μM, and a respective minimum bactericidal concentration (MBC) of 12.5 μM and 25 μM. However, the MIC and MBC values of DHA or EPA against F. nucleatum were both greater than 100 μM. For early-stage bacteria, DHA or EPA displayed complete inhibition on the planktonic growth and biofilm formation of P. gingivalis from the lowest concentration of 12.5 μM. And the planktonic growth of F. nucleatum was slightly but not completely inhibited by DHA or EPA even at the concentration of 100 μM, however, the biofilm formation of F. nucleatum at 24 h was significantly restrained by 100 μM EPA. For exponential-phase bacteria, 100 μM DHA or EPA completely killed P. gingivalis and significantly decreased the viable counts of F. nucleatum. Meanwhile, the morphology of P. gingivalis was apparently damaged, and the virulence factor gene expression of P. gingivalis and F. nucleatum was strongly downregulated. Besides, the viability and the thickness of mature P. gingivalis biofilm, together with the viability of mature F. nucleatum biofilm were both significantly decreased in the presence of 100 μM DHA or EPA. In conclusion, DHA and EPA possessed antibacterial activities against planktonic and biofilm forms of periodontal pathogens, which suggested that DHA and EPA might be potentially supplementary therapeutic agents for prevention and treatment of periodontal diseases.
Collapse
Affiliation(s)
- Mengjun Sun
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Zichao Zhou
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Jiachen Dong
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Jichun Zhang
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Yiru Xia
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China
| | - Rong Shu
- Department of Periodontology, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Key Laboratory of Stomatology, Shanghai 200011, China.
| |
Collapse
|
18
|
Jung SW, Lee SW. The antibacterial effect of fatty acids on Helicobacter pylori infection. Korean J Intern Med 2016; 31:30-5. [PMID: 26767854 PMCID: PMC4712431 DOI: 10.3904/kjim.2016.31.1.30] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 12/09/2015] [Indexed: 02/06/2023] Open
Abstract
Eradication of Helicobacter pylori is recommended for the management of various gastric diseases, including peptic ulcers and mucosa-associated lymphoid tissue lymphoma. Because of the increasing prevalence of antibiotic resistance, the eradication rates of antibiotic-based therapies have decreased. Therefore, alternative treatments should be considered. The antibacterial properties of fatty acids (FAs) have been investigated in various organisms, including H. pylori. Some FAs, particularly polyunsaturated FAs, have been shown to have bactericidal activity against H. pylori in vitro; however, their antibacterial effects in vivo remain controversial. Poor solubility and delivery of FAs may be important reasons for this discrepancy. Recently, a series of studies demonstrated the antibacterial effects of a liposomal formulation of linolenic acid against H. pylori, both in vitro and in vivo. Further research is needed to improve the bioavailability of FAs and apply them in clinical use.
Collapse
Affiliation(s)
- Sung Woo Jung
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
| | - Sang Woo Lee
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Korea
- Correspondence to Sang Woo Lee, M.D. Department of Internal Medicine, Institute of Digestive Disease and Nutrition, Korea University Ansan Hospital, 123 Jeokgeum-ro, Danwon-gu, Ansan 15355, Korea Tel: +82-31-412-5580 Fax: +82-31-8099-6373 E-mail:
| |
Collapse
|
19
|
Omega-3 Polyunsaturated Fatty Acids Intake to Regulate Helicobacter pylori-Associated Gastric Diseases as Nonantimicrobial Dietary Approach. BIOMED RESEARCH INTERNATIONAL 2015; 2015:712363. [PMID: 26339635 PMCID: PMC4538587 DOI: 10.1155/2015/712363] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 12/20/2014] [Accepted: 01/15/2015] [Indexed: 02/07/2023]
Abstract
Omega-3 polyunsaturated fatty acids (n-3 PUFAs), commonly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), have been acknowledged as essential long-chain fatty acids imposing either optimal health promotion or the rescuing from chronic inflammatory diseases such as atherosclerosis, fatty liver, and various inflammatory gastrointestinal diseases. Recent studies dealing with EPA and DHA have sparked highest interests because detailed molecular mechanisms had been documented with the identification of its receptor, G protein coupled receptor, and GPR120. In this review article, we have described clear evidences showing that n-3 PUFAs could reduce various Helicobacter pylori- (H. pylori-) associated gastric diseases and extended to play even cancer preventive outcomes including H. pylori-associated gastric cancer by influencing multiple targets, including proliferation, survival, angiogenesis, inflammation, and metastasis. Since our previous studies strongly concluded that nonantimicrobial dietary approach for reducing inflammation, for instance, application of phytoceuticals, probiotics, natural products including Korean red ginseng, and walnut plentiful of n-3 PUFAs, might be prerequisite step for preventing H. pylori-associated gastric cancer as well as facilitating the rejuvenation of precancerous atrophic gastritis, these beneficial lipids can restore or modify inflammation-associated lipid distortion and correction of altered lipid rafts to send right signaling to maintain healthy stomach even after chronic H. pylori infection.
Collapse
|
20
|
Yamashita S, Igarashi M, Hayashi C, Shitara T, Nomoto A, Mizote T, Shibasaki M. Identification of self-growth-inhibiting compounds lauric acid and 7-(Z)-tetradecenoic acid from Helicobacter pylori. MICROBIOLOGY-SGM 2015; 161:1231-9. [PMID: 25767109 DOI: 10.1099/mic.0.000077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Helicobacter pylori growth medium is usually supplemented with horse serum (HS) or FCS. However, cyclodextrin derivatives or activated charcoal can replace serum. In this study, we purified self-growth-inhibiting (SGI) compounds from H. pylori growth medium. The compounds were recovered from porous resin, Diaion HP-20, which was added to the H. pylori growth medium instead of known supplements. These SGI compounds were also identified from 2,6-di-O-methyl-β-cyclodextrin, which was supplemented in a pleuropneumonia-like organisms broth. The growth-inhibiting compounds were identified as lauric acid (LA) and 7-(Z)-tetradecenoic acid [7-(Z)-TDA]. Although several fatty acids had been identified in H. pylori, these specific compounds were not previously found in this species. However, we confirmed that these fatty acids were universally present in the cultivation medium of the H. pylori strains examined in this study. A live/dead assay carried out without HS indicated that these compounds were bacteriostatic; however, no significant growth-inhibiting effect was observed against other tested bacterial species that constituted the indigenous bacterial flora. These findings suggested that LA and 7-(Z)-TDA might play important roles in the survival of H. pylori in human stomach epithelial cells.
Collapse
Affiliation(s)
- Shinpei Yamashita
- 1Department of Human Nutrition, Yamaguchi Prefectural University, Yamaguchi, Japan
| | | | | | - Tetsuo Shitara
- 2Institute of Microbial Chemistry (BIKAKEN), Tokyo, Japan
| | - Akio Nomoto
- 2Institute of Microbial Chemistry (BIKAKEN), Tokyo, Japan
| | - Tomoko Mizote
- 1Department of Human Nutrition, Yamaguchi Prefectural University, Yamaguchi, Japan
| | | |
Collapse
|
21
|
Helicobacter pylori's cholesterol uptake impacts resistance to docosahexaenoic acid. Int J Med Microbiol 2014; 304:314-20. [DOI: 10.1016/j.ijmm.2013.11.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 11/06/2013] [Accepted: 11/25/2013] [Indexed: 12/14/2022] Open
|