1
|
Sun W, Zhang C, Xu J, Zhao M, Li P. Natural small-molecule compounds targeting Helicobacter pylori virulence factors: A promising strategy for overcoming antibiotic resistance. Biochem Biophys Res Commun 2025; 768:151877. [PMID: 40334425 DOI: 10.1016/j.bbrc.2025.151877] [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: 02/10/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/09/2025]
Abstract
Helicobacter pylori (H. pylori) infection is an important causal factor of gastritis, peptic ulcer, and gastric cancer. High infection rates and the increasing challenge of antibiotic resistance worldwide have prompted an urgent need to develop novel therapeutic options and antimicrobial agents. This review focuses on the potential of natural small-molecule compounds as novel anti-H. pylori agents-a promising approach that mitigates the risk of resistance development and maintains the microbiome's ecological balance. We detail how H. pylori virulence factors, including urease, CagA, VacA, and biofilm, contribute to pathogenicity and underline the reassuring fact that naturally derived compounds sourced from plants and microorganisms have shown remarkable efficacy in inhibiting these virulence factors. Some compounds also exhibit synergistic effects with conventional antibiotics, potentially overcoming challenges associated with resistant strains. Furthermore, we discuss recent advancements in identifying novel drug targets within the H. pylori virulence spectrum, offering insights into future directions for research and development in H. pylori therapy.
Collapse
Affiliation(s)
- Wenjing Sun
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China; State Key Laboratory of Digestive Health, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing, 100050, China
| | - Congen Zhang
- Department of Pharmacy, Beijing Friendship Hospital, Capital Medical University, 100050 Beijing, China
| | - Junxuan Xu
- State Key Laboratory of Digestive Health, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing, 100050, China.
| | - Mengran Zhao
- State Key Laboratory of Digestive Health, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing, 100050, China.
| | - Peng Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang, China; State Key Laboratory of Digestive Health, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing, 100050, China.
| |
Collapse
|
2
|
Unakal C, Blake LW, Farfan G, Justiz-Vaillant A, Pandit BR, Akpaka PE. Helicobacter pylori vacA genes associated with gastric diseases in Trinidad and Tobago. IJID REGIONS 2025; 14:100498. [PMID: 39717866 PMCID: PMC11664410 DOI: 10.1016/j.ijregi.2024.100498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 12/25/2024]
Abstract
Objectives To delineate and understand the genetic variations among Helicobacter pylori strains from Trinidad and Tobago associated with gastric diseases. Methods One hundred (n = 100) patients who routinely presented with clinical features suggestive of peptic disease were enrolled in the study and underwent gastroscopy procedures. Biopsy specimens were analyzed using serological and molecular methods. Amplification of the variable vacuolating cytotoxin gene A (vacA), urease C (ureC), and 16S ribosomal RNA regions was performed by polymerase chain reaction; and then followed by sequencing and alignment. Phylogenetic analysis and DNA fragment alignment were carried out using DNA MEGA 7 version 7. Results Overall, 70 of the patients tested positive for H. pylori infection using an enzyme immune assay. All were 16S ribosomal RNA positive, with 8.6% (6/70) testing positive for S1 and 91.4% (64/70) for S2. The most prevalent vacA allelic combination was S2M1, with 87.1% (61/70) cases. All S1M1 strains were ureC-positive, while S1M2 strains were ureC-negative. Conclusions H. pylori strain variations are seen in the Trinidad and Tobago isolates and are most closely related to the ones from the USA, India, and Indonesia. However, the factors contributing to the emergence of S2/M1 being more prevalent will need to be further investigated.
Collapse
Affiliation(s)
- Chandrashekhar Unakal
- Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Lemar Wayne Blake
- Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Gerard Farfan
- The Endoscopy Centre, Woodbrook, Port of Spain, Trinidad and Tobago
| | - Angel Justiz-Vaillant
- Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Bijay Raj Pandit
- Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| | - Patrick Eberechi Akpaka
- Faculty of Medical Sciences, The University of the West Indies, St. Augustine, Trinidad and Tobago
| |
Collapse
|
3
|
Huang Y, Wang X, Liu H, Meng X, Yin H, Hou R, Lin W, Zhang X, Ma J, Zhang X, Zhang F, Miao Y. Knocking Down HN1 Blocks Helicobacter pylori-Induced Malignant Phenotypes in Gastric Mucosal Cells and Inhibits Gastric Cancer Cell Proliferation, Cytoskeleton Remodeling, and Migration. Biochem Genet 2025; 63:875-895. [PMID: 38526710 DOI: 10.1007/s10528-024-10731-7] [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: 11/14/2023] [Accepted: 02/02/2024] [Indexed: 03/27/2024]
Abstract
Helicobacter pylori (H. pylori) is implicated in the aberrant proliferation and malignant transformation of gastric mucosal cells, heightening the risk of gastric cancer (GC). HN1 is involved in the development of various tumors. However, precise mechanistic underpinnings of HN1 promoting GC progression in H. pylori remain elusive. The study collected 79 tissue samples of GC patients, including 47 with H. pylori-positive GC and 32 H. pylori-negative controls. Using human gastric epithelial cells (GES-1) and human gastric adenocarcinoma cells (HGC-27), the effect of overexpression / knocking down of HN1 and H. pylori infection was evaluated on cell function (proliferation, migration, apoptosis), cytoskeleton, and expression of cell malignant phenotype factors that promote the malignant biological behavior of cancer cells. The expression of HN1 in GC tissues is higher than that in paracancerous tissue and is closely related to infiltration, lymphatic metastasis, distant metastasis, survival, and H. pylori infection. Downregulation of HN1 effectively hinders the ability of H. pylori strains 26695 and SS1 to promote migration of GES-1 and HGC-27 cells, while lowering the expression of key indicators associated with malignant phenotype. Downregulated GSK3B, β-catenin, and Vimentin after knockdown Integrinβ1, but HN1 expression remained largely unchanged, when HN1 and Integrinβ1 were knocked down, GSK3B, β-catenin, and Vimentin expression were considerably reduced. Our research demonstrated the crucial role of HN1 in H. pylori-induced acquisition of a malignant phenotype in GES-1 cells. Knockdown of HN1 blocked the pathogenic mechanism of H. pylori-induced GC and downregulated the expression of GSK3Β, β-catenin and Vimentin via Integrin β1.
Collapse
Affiliation(s)
- Ying Huang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Hui Autonomous Region, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Xiaofei Wang
- Department of Pathology, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000, Hebie Province, China
| | - Hao Liu
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Hui Autonomous Region, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Xiangkun Meng
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Hui Autonomous Region, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Hua Yin
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Hui Autonomous Region, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Ruirui Hou
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Hui Autonomous Region, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Wan Lin
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Hui Autonomous Region, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Xu Zhang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Hui Autonomous Region, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Jun Ma
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Hui Autonomous Region, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Xiaoxu Zhang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Hui Autonomous Region, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Feixiong Zhang
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Hui Autonomous Region, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China
| | - Yu Miao
- Department of Gastroenterology, General Hospital of Ningxia Medical University, Hui Autonomous Region, 804 Shengli South Street, Xingqing District, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
4
|
Budriesi R, Corazza I, Roncioni S, Scanferlato R, De Luca D, Marzetti C, Gotti R, Rizzardi N, Bergamini C, Micucci M, Roncarati D, Mattioli LB. Herbal Extracts Mixed with Essential Oils: A Network Approach for Gastric and Intestinal Motility Disorders. Nutrients 2024; 16:4357. [PMID: 39770978 PMCID: PMC11677010 DOI: 10.3390/nu16244357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Three herbal extracts (Asparagus racemosus Willd., Tabebuia avellanedae Lorentz, and Glycyrrhiza glabra L.) were mixed with three essential oils (Foeniculum vulgare Mill., Mentha piperita L., and Pimpinella anisum L.) to formulate a product (HEMEO) whose active compounds include saponins and steroids in Asparagus racemosus, known for their anti-inflammatory properties; glycyrrhizin and flavonoids in Glycyrrhiza glabra, which exhibit gastroprotective and antispasmodic effects; menthol in Mentha piperita, contributing with antispasmodic and antimicrobial properties; and anethole and polyphenols in Pimpinella anisum, which modulate intestinal motility and offer antimicrobial activity. OBJECTIVE HEMEO was formulated for applications in intestinal motility disorders. METHODS HEMEO was evaluated for spontaneous and induced motility effects in isolated guinea pig ileum, colon, and stomach. Ex vivo experiments were conducted using LabChart software v7.0, and the product's antibacterial action against Helicobacter pylori and its antioxidant effects were assessed through disc diffusion and FRAP assays. The presence of the volatile compounds in the formulation was confirmed by GC-MS analysis; the TPC of HEMEO, determined using the Folin-Ciocalteu method, was 9.925 ± 0.42 mg GAE/g. CONCLUSIONS HEMEO showed a phenolic content correlated with its antioxidant potential and in addition inhibited H. pylori growth and demonstrated notable antioxidant properties, suggesting its role as a supportive agent in digestive processes and in managing motility disorders.
Collapse
Affiliation(s)
- Roberta Budriesi
- Food Chemistry and Nutraceutical Laboratory, Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy;
| | - Ivan Corazza
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum-University of Bologna, 40138 Bologna, Italy;
| | - Simone Roncioni
- Valsambro S.r.l., 40121 Bologna, Italy; (S.R.); (R.S.); (D.D.L.); (C.M.)
| | | | - Dalila De Luca
- Valsambro S.r.l., 40121 Bologna, Italy; (S.R.); (R.S.); (D.D.L.); (C.M.)
| | - Carla Marzetti
- Valsambro S.r.l., 40121 Bologna, Italy; (S.R.); (R.S.); (D.D.L.); (C.M.)
| | - Roberto Gotti
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Nicola Rizzardi
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (N.R.); (C.B.)
| | - Christian Bergamini
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Irnerio 48, 40126 Bologna, Italy; (N.R.); (C.B.)
| | - Matteo Micucci
- Department of Biomolecular Sciences, University of Urbino “Carlo Bo”, 61029 Urbino, Italy;
| | - Davide Roncarati
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, Via Selmi 3, 40126 Bologna, Italy;
| | - Laura Beatrice Mattioli
- Food Chemistry and Nutraceutical Laboratory, Department of Pharmacy and Biotechnology (FaBiT), Alma Mater Studiorum-University of Bologna, Via Belmeloro 6, 40126 Bologna, Italy;
| |
Collapse
|
5
|
Kacemi R, Campos MG. Bee Pollen as a Source of Biopharmaceuticals for Neurodegeneration and Cancer Research: A Scoping Review and Translational Prospects. Molecules 2024; 29:5893. [PMID: 39769981 PMCID: PMC11677910 DOI: 10.3390/molecules29245893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/18/2024] [Accepted: 11/20/2024] [Indexed: 01/03/2025] Open
Abstract
Bee Pollen (BP) has many advantageous properties relying on its multitargeting potential, a new tendency in managing many challenging illnesses. In cancer and neurodegeneration, the multiple effects of BP could be of unequaled importance and need further investigation. Although still limited, available data interestingly spotlights some floral sources with promising activities in line with this investigation. Adopting scoping review methodology, we have identified many crucial bioactivities that are widely recognized to individual BP compounds but remain completely untapped in this valuable bee cocktail. A wide range of these compounds have been recently found to be endowed with great potential in modulating pivotal processes in neurodegeneration and cancer pathophysiology. In addition, some ubiquitous BP compounds have only been recently isolated, while the number of studied BPs remains extremely limited compared to the endless pool of plant species worldwide. We have also elucidated that clinical profits from these promising perspectives are still impeded by challenging hurdles such as limited bioavailability of the studied phytocompounds, diversity and lack of phytochemical standardization of BP, and the difficulty of selective targeting in some pathophysiological mechanisms. We finally present interesting insights to guide future research and pave the way for urgently needed and simplified clinical investigations.
Collapse
Affiliation(s)
- Rachid Kacemi
- Observatory of Drug-Herb Interactions, Faculty of Pharmacy, Heath Sciences Campus, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
| | - Maria G. Campos
- Observatory of Drug-Herb Interactions, Faculty of Pharmacy, Heath Sciences Campus, University of Coimbra, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal;
- Coimbra Chemistry Centre (CQC, FCT Unit 313) (FCTUC), University of Coimbra, Rua Larga, 3004-531 Coimbra, Portugal
| |
Collapse
|
6
|
Elbehiry A, Abalkhail A, Anajirih N, Alkhamisi F, Aldamegh M, Alramzi A, AlShaqi R, Alotaibi N, Aljuaid A, Alzahrani H, Alzaben F, Rawway M, Ibrahem M, Abdelsalam MH, Rizk NI, Mostafa MEA, Alfaqir MR, Edrees HM, Alqahtani M. Helicobacter pylori: Routes of Infection, Antimicrobial Resistance, and Alternative Therapies as a Means to Develop Infection Control. Diseases 2024; 12:311. [PMID: 39727641 PMCID: PMC11727528 DOI: 10.3390/diseases12120311] [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: 10/14/2024] [Revised: 11/16/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024] Open
Abstract
Helicobacter pylori (H. pylori) is a Gram-negative, spiral-shaped bacterium that colonizes the gastric epithelium and is associated with a range of gastrointestinal disorders, exhibiting a global prevalence of approximately 50%. Despite the availability of treatment options, H. pylori frequently reemerges and demonstrates increasing antibiotic resistance, which diminishes the efficacy of conventional therapies. Consequently, it is imperative to explore non-antibiotic treatment alternatives to mitigate the inappropriate use of antibiotics. This review examines H. pylori infection, encompassing transmission pathways, treatment modalities, antibiotic resistance, and eradication strategies. Additionally, it discusses alternative therapeutic approaches such as probiotics, anti-biofilm agents, phytotherapy, phototherapy, phage therapy, lactoferrin therapy, and vaccine development. These strategies aim to reduce antimicrobial resistance and enhance treatment outcomes for H. pylori infections. While alternative therapies can maintain low bacterial levels, they do not achieve complete eradication of H. pylori. These therapies are designed to bolster the immune response, minimize side effects, and provide gastroprotective benefits, rendering them suitable for adjunctive use alongside conventional treatments. Probiotics may serve as adjunctive therapy for H. pylori; however, their effectiveness as a monotherapy is limited. Photodynamic and phage therapies exhibit potential in targeting H. pylori infections, including those caused by drug-resistant strains, without the use of antibiotics. The development of a reliable vaccine is also critical for the eradication of H. pylori. This review identifies candidate antigens such as VacA, CagA, and HspA, along with various vaccine formulations, including vector-based and subunit vaccines. Some vaccines have demonstrated efficacy in clinical trials, while others have shown robust immune protection in preclinical studies. Nevertheless, each of the aforementioned alternative therapies requires thorough preclinical and clinical evaluation to ascertain their efficacy, side effects, cost-effectiveness, and patient compliance.
Collapse
Affiliation(s)
- Ayman Elbehiry
- Department of Public Health, College of Applied Medical Sciences, Qassim University, P.O. Box 6666, Buraydah 51452, Saudi Arabia
| | - Adil Abalkhail
- Department of Public Health, College of Applied Medical Sciences, Qassim University, P.O. Box 6666, Buraydah 51452, Saudi Arabia
| | - Nuha Anajirih
- Medical Emergency Services Department, Faculty of Health Sciences, Umm Al-Qura University, Al-Qunfudah P.O. Box 1109, Saudi Arabia
| | - Fahad Alkhamisi
- Department of Preventive Medicine, King Fahad Armed Hospital, Jeddah 23311, Saudi Arabia
| | - Mohammed Aldamegh
- Pathology and Laboratory Medicine Department, Armed Forces Hospital-Jubail, Jubail 31951, Saudi Arabia
| | - Abdullah Alramzi
- Medical Radiology Department, Armed Forces Hospital-Jubail, Jubail 31951, Saudi Arabia
| | - Riyad AlShaqi
- Biomedical Engineer, Armed Forces Medical Services, Riyadh 12426, Saudi Arabia
| | - Naif Alotaibi
- Medical Hospital Administration Department, Armed Forces Hospital-Jubail, Jubail 31951, Saudi Arabia
| | - Abdullah Aljuaid
- Medical Hospital Administration Department, Armed Forces Hospitals in Al Kharj, AL Kharj 16278, Saudi Arabia
| | - Hilal Alzahrani
- Physical Medicine and Rehabilitation Department, Armed Forces Center for Health Rehabilitation, Taif 21944, Saudi Arabia
| | - Feras Alzaben
- Department of Food Service, King Fahad Armed Forces Hospital, Jeddah 23311, Saudi Arabia
| | - Mohammed Rawway
- Biology Department, College of Science, Jouf University, Sakaka 42421, Saudi Arabia
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Assiut 71524, Egypt
| | - Mai Ibrahem
- Department of Public Health, College of Applied Medical Science, King Khalid University, Abha 61421, Saudi Arabia
| | - Moustafa H. Abdelsalam
- Department of Physiology, Faculty of Medicine, University of Tabuk, Tabuk 74191, Saudi Arabia
| | - Nermin I. Rizk
- Department of Physiology, Faculty of Medicine, University of Tabuk, Tabuk 74191, Saudi Arabia
| | - Mohamed E. A. Mostafa
- Department of Anatomy, Faculty of Medicine, University of Tabuk, Tabuk 74191, Saudi Arabia
| | - Moneef Rohail Alfaqir
- Department of Anatomy, Faculty of Medicine, University of Tabuk, Tabuk 74191, Saudi Arabia
| | - Husam M. Edrees
- Department of Physiology, Faculty of Medicine, University of Tabuk, Tabuk 74191, Saudi Arabia
| | - Mubarak Alqahtani
- Department of Radiology, King Fahd Armed Forces Hospital, Jeddah 23311, Saudi Arabia
| |
Collapse
|
7
|
Chauhan A, Yadav M, Chauhan R, Basniwal RK, Pathak VM, Ranjan A, Kapardar RK, Srivastav R, Tuli HS, Ramniwas S, Mathkor DM, Haque S, Hussain A. Exploring the Potential of Ellagic Acid in Gastrointestinal Cancer Prevention: Recent Advances and Future Directions. Oncol Ther 2024; 12:685-699. [PMID: 39222186 PMCID: PMC11574235 DOI: 10.1007/s40487-024-00296-1] [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: 05/13/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Gastrointestinal (GI) cancers are a significant global health concern with diverse etiologies and limited treatment options. Ellagic acid (EA), a natural polyphenolic compound, exhibits promising anticancer properties against various GI malignancies. In this article, we have reviewed recent research on the anticancer potential of EA across esophageal, gastric, colorectal, pancreatic, and liver cancers. In esophageal cancer, EA inhibits the formation of O6-methylguanine (O6-meGua) adducts induced by carcinogens like N-nitrosomethylbenzylamine (NMBA), thereby suppressing tumor growth. Additionally, EA inhibits STAT3 signaling and stabilizes tumor suppressor proteins, showing potential as an anti-esophageal cancer agent. In gastric cancer, EA regulates multiple pathways involved in cell proliferation, invasion, and apoptosis, including the p53 and PI3K-Akt signaling pathways. It also demonstrates anti-inflammatory and antioxidant effects, making it a promising therapeutic candidate against gastric cancer. In colorectal cancer (CRC), EA inhibits cell proliferation, induces apoptosis, and modulates the Wnt/β-catenin and PI3K/Akt pathways, suggesting its efficacy in preventing CRC progression. Furthermore, EA has shown promise in pancreatic cancer by inhibiting nuclear factor-kappa B, inducing apoptosis, and suppressing epithelial-mesenchymal transition. In liver cancer, EA exhibits radio-sensitizing effects, inhibits inflammatory pathways, and modulates the tumor microenvironment, offering potential therapeutic benefits against hepatocellular carcinoma. Studies on EA potential in combination therapies and the development of targeted delivery systems are required for enhanced efficacy against gastrointestinal cancers.
Collapse
Affiliation(s)
- Abhishek Chauhan
- Amity Institute of Environmental Toxicology Safety and Management, Amity University, Noida, U.P., India
| | - Monika Yadav
- Cancer Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Ritu Chauhan
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, 248002, India
| | - Rupesh Kumar Basniwal
- Amity Institute of Advanced Research and Studies (M&D), Amity University, Noida, U.P., India
| | - Vinay Mohan Pathak
- Parwatiya Shiksha Sabha (PASS), Near Transport Nagar Develchaur Kham, Haldwani, Nainital, India
| | - Anuj Ranjan
- Academy of Biology and Biotechnology, Southern Federal University, Stachki 194/1, Rostov-on-Don, 344090, Russia
| | | | - Rajpal Srivastav
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Noida, India
| | - Hardeep Singh Tuli
- Department of Biosciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - Seema Ramniwas
- University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali, 140413, India
| | - Darin Mansor Mathkor
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, 45142, Jazan, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, 45142, Jazan, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, 11022801, Lebanon
| | - Arif Hussain
- School of Life Sciences, Manipal Academy of Higher Education, P.O. Box 345050, Dubai, United Arab Emirates.
| |
Collapse
|
8
|
Aborabu AAS, Tayel AA, Assas M, Moussa SH, Alalawy AI, Almutairi FM, Omar AA. Anti-Helicobacter pylori activity of nanocomposites from chitosan/broccoli mucilage/selenium nanoparticles. Sci Rep 2024; 14:21693. [PMID: 39289449 PMCID: PMC11408496 DOI: 10.1038/s41598-024-65762-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 06/24/2024] [Indexed: 09/19/2024] Open
Abstract
Helicobacter pylori can infect most people worldwide to cause hazardous consequences to health; the bacteria could not easily be controlled or disinfected. Toward exploring of innovative biocidal nanoformulations to control H. pylori, broccoli seeds (Brassica oleracea var. italica) mucilage (MBS) was employed for biosynthesizing selenium nanoparticles (MBS/SeNPs), which was intermingled with chitosan nanoparticles (NCT) to generate bioactive nanocomposites for suppressing H. pylori. The MBS could effectually generate and stabilize SeNPs with 13.61 nm mean diameter, where NCT had 338.52 nm mean diameter and positively charged (+ 39.62 mV). The cross-linkages between NCT-MBS-SeNPs were verified via infrared analysis and the nanocomposites from NCT:MBS/SeNPs at 1:2 (T1), 1:1 (T2) and 2:1 (T3) ratios had mean diameters of 204, 132 and 159 nm, respectively. The entire nanomaterials/composites exhibited potent anti- H. pylori activities using various assaying methods; the T2 nanocomposite was the utmost bactericidal agent with 0.08-0.10 mg/L minimal concentration and 25.9-27.3 mm inhibition zones. The scanning microscopy displayed the ability of nanocomposite to attach the bacterial cells, disrupt their membranes, and completely lyse them within 10 h. The NCT/MBS/SeNPs nanocomposites provided effectual innovative approach to control H. pylori.
Collapse
Affiliation(s)
- Ahlam A S Aborabu
- Department of Fish Processing and Biotechnology, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Ahmed A Tayel
- Department of Fish Processing and Biotechnology, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt.
- Academy of Scientific Research & Technology (ASRT), Cairo, 11516, Egypt.
| | - Mona Assas
- Department of Fish Processing and Biotechnology, Faculty of Aquatic and Fisheries Sciences, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| | - Shaaban H Moussa
- Department of Biology, College of Science and Humanitarian Studies, Shaqra University, 11961, Shaqra, Saudi Arabia.
| | - Adel I Alalawy
- Biochemistry Department, Faculty of Science, University of Tabuk, 47512, Tabuk, Saudi Arabia
| | - Fahad M Almutairi
- Biochemistry Department, Faculty of Science, University of Tabuk, 47512, Tabuk, Saudi Arabia
| | - Amira A Omar
- Department of Fish Diseases and Management, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, 33516, Egypt
| |
Collapse
|
9
|
Angelini P. Plant-Derived Antimicrobials and Their Crucial Role in Combating Antimicrobial Resistance. Antibiotics (Basel) 2024; 13:746. [PMID: 39200046 PMCID: PMC11350763 DOI: 10.3390/antibiotics13080746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 07/27/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024] Open
Abstract
Antibiotic resistance emerged shortly after the discovery of the first antibiotic and has remained a critical public health issue ever since. Managing antibiotic resistance in clinical settings continues to be challenging, particularly with the rise of superbugs, or bacteria resistant to multiple antibiotics, known as multidrug-resistant (MDR) bacteria. This rapid development of resistance has compelled researchers to continuously seek new antimicrobial agents to curb resistance, despite a shrinking pipeline of new drugs. Recently, the focus of antimicrobial discovery has shifted to plants, fungi, lichens, endophytes, and various marine sources, such as seaweeds, corals, and other microorganisms, due to their promising properties. For this review, an extensive search was conducted across multiple scientific databases, including PubMed, Elsevier, ResearchGate, Scopus, and Google Scholar, encompassing publications from 1929 to 2024. This review provides a concise overview of the mechanisms employed by bacteria to develop antibiotic resistance, followed by an in-depth exploration of plant secondary metabolites as a potential solution to MDR pathogens. In recent years, the interest in plant-based medicines has surged, driven by their advantageous properties. However, additional research is essential to fully understand the mechanisms of action and verify the safety of antimicrobial phytochemicals. Future prospects for enhancing the use of plant secondary metabolites in combating antibiotic-resistant pathogens will also be discussed.
Collapse
Affiliation(s)
- Paola Angelini
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06122 Perugia, Italy
| |
Collapse
|
10
|
Boyanova L, Medeiros J, Yordanov D, Gergova R, Markovska R. Turmeric and curcumin as adjuncts in controlling Helicobacter pylori-associated diseases: a narrative review. Lett Appl Microbiol 2024; 77:ovae049. [PMID: 38794899 DOI: 10.1093/lambio/ovae049] [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: 03/06/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 05/26/2024]
Abstract
Non-antibiotic adjuncts may improve Helicobacter pylori infection control. Our aim was to emphasize curcumin benefits in controlling H. pylori infection. We discussed publications in English mostly published since 2020 using keyword search. Curcumin is the main bioactive substance in turmeric. Curcumin inhibited H. pylori growth, urease activity, three cag genes, and biofilms through dose- and strain-dependent activities. Curcumin also displayed numerous anticancer activities such as apoptosis induction, anti-inflammatory and anti-angiogenic effects, caspase-3 upregulation, Bax protein enhancement, p53 gene activation, and chemosensitization. Supplementing triple regimens, the agent increased H. pylori eradication success in three Iranian studies. Bioavailability was improved by liposomal preparations, lipid conjugates, electrospray-encapsulation, and nano-complexation with proteins. The agent was safe at doses of 0.5->4 g daily, the most common (in 16% of the users) adverse effect being gastrointestinal upset. Notably, curcumin favorably influences the intestinal microbiota and inhibits Clostridioides difficile. Previous reports showed the inhibitory effect of curcumin on H pylori growth. Curcumin may become an additive in the therapy of H. pylori infection, an adjunct for gastric cancer control, and an agent beneficial to the intestinal microbiota. Further examination is necessary to determine its optimal dosage, synergy with antibiotics, supplementation to various eradication regimens, and prophylactic potential.
Collapse
Affiliation(s)
- Lyudmila Boyanova
- Department of Medical Microbiology, Medical University of Sofia, 1431 Sofia, Bulgaria
| | - José Medeiros
- Gastroenterology Clinic, Rua do Carmo, 75-1º AA,, 3000 Coimbra, Portugal
| | - Daniel Yordanov
- Department of Medical Microbiology, Medical University of Sofia, 1431 Sofia, Bulgaria
| | - Raina Gergova
- Department of Medical Microbiology, Medical University of Sofia, 1431 Sofia, Bulgaria
| | - Rumyana Markovska
- Department of Medical Microbiology, Medical University of Sofia, 1431 Sofia, Bulgaria
| |
Collapse
|
11
|
Morin C, Verma VT, Arya T, Casu B, Jolicoeur E, Ruel R, Marinier A, Sygusch J, Baron C. Structure-based design of small molecule inhibitors of the cagT4SS ATPase Cagα of Helicobacter pylori. Biochem Cell Biol 2024; 102:226-237. [PMID: 38377487 DOI: 10.1139/bcb-2023-0331] [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] [Indexed: 02/22/2024] Open
Abstract
We here describe the structure-based design of small molecule inhibitors of the type IV secretion system of Helicobacter pylori. The secretion system is encoded by the cag pathogenicity island, and we chose Cagα, a hexameric ATPase and member of the family of VirB11-like proteins, as target for inhibitor design. We first solved the crystal structure of Cagα in a complex with the previously identified small molecule inhibitor 1G2. The molecule binds at the interface between two Cagα subunits and mutagenesis of the binding site identified Cagα residues F39 and R73 as critical for 1G2 binding. Based on the inhibitor binding site we synthesized 98 small molecule derivates of 1G2 to improve binding of the inhibitor. We used the production of interleukin-8 of gastric cancer cells during H. pylori infection to screen the potency of inhibitors and we identified five molecules (1G2_1313, 1G2_1338, 1G2_2886, 1G2_2889, and 1G2_2902) that have similar or higher potency than 1G2. Differential scanning fluorimetry suggested that these five molecules bind Cagα, and enzyme assays demonstrated that some are more potent ATPase inhibitors than 1G2. Finally, scanning electron microscopy revealed that 1G2 and its derivatives inhibit the assembly of T4SS-determined extracellular pili suggesting a mechanism for their anti-virulence effect.
Collapse
Affiliation(s)
- Claire Morin
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Québec, Canada
| | - Vijay Tailor Verma
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Québec, Canada
| | - Tarun Arya
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Québec, Canada
| | - Bastien Casu
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Québec, Canada
| | - Eric Jolicoeur
- Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Québec, Canada
| | - Réjean Ruel
- Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Québec, Canada
| | - Anne Marinier
- Institut de Recherche en Immunologie et Cancérologie, Université de Montréal, Québec, Canada
- Department of Chemistry, Faculty of Arts and Sciences, Université de Montréal, Québec, Canada
| | - Jurgen Sygusch
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Québec, Canada
| | - Christian Baron
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Québec, Canada
| |
Collapse
|
12
|
Vieira RV, Peiter GC, de Melo FF, Zarpelon-Schutz AC, Teixeira KN. In silico prospective analysis of the medicinal plants activity on the CagA oncoprotein from Helicobacter pylori. World J Clin Oncol 2024; 15:653-663. [PMID: 38835850 PMCID: PMC11145963 DOI: 10.5306/wjco.v15.i5.653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/01/2024] [Accepted: 04/18/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND Colonization with Helicobacter pylori (H. pylori) has a strong correlation with gastric cancer, and the virulence factor CagA is implicated in carcinogenesis. Studies have been conducted using medicinal plants with the aim of eliminating the pathogen; however, the possibility of blocking H. pylori-induced cell differentiation to prevent the onset and/or progression of tumors has not been addressed. This type of study is expensive and time-consuming, requiring in vitro and/or in vivo tests, which can be solved using bioinformatics. Therefore, prospective computational analyses were conducted to assess the feasibility of interaction between phenolic compounds from medicinal plants and the CagA oncoprotein. AIM To perform a computational prospecting of the interactions between phenolic compounds from medicinal plants and the CagA oncoprotein of H. pylori. METHODS In this in silico study, the structures of the phenolic compounds (ligands) kaempferol, myricetin, quercetin, ponciretin (flavonoids), and chlorogenic acid (phenolic acid) were selected from the PubChem database. These phenolic compounds were chosen based on previous studies that suggested medicinal plants as non-drug treatments to eliminate H. pylori infection. The three-dimensional structure model of the CagA oncoprotein of H. pylori (receptor) was obtained through molecular modeling using computational tools from the I-Tasser platform, employing the threading methodology. The primary sequence of CagA was sourced from GenBank (BAK52797.1). A screening was conducted to identify binding sites in the structure of the CagA oncoprotein that could potentially interact with the ligands, utilizing the GRaSP online platform. Both the ligands and receptor were prepared for molecular docking using AutoDock Tools 4 (ADT) software, and the simulations were carried out using a combination of ADT and AutoDock Vina v.1.2.0 software. Two sets of simulations were performed: One involving the central region of CagA with phenolic compounds, and another involving the carboxy-terminus region of CagA with phenolic compounds. The receptor-ligand complexes were then analyzed using PyMol and BIOVIA Discovery Studio software. RESULTS The structure model obtained for the CagA oncoprotein exhibited high quality (C-score = 0.09) and was validated using parameters from the MolProbity platform. The GRaSP online platform identified 24 residues (phenylalanine and leucine) as potential binding sites on the CagA oncoprotein. Molecular docking simulations were conducted with the three-dimensional model of the CagA oncoprotein. No complexes were observed in the simulations between the carboxy-terminus region of CagA and the phenolic compounds; however, all phenolic compounds interacted with the central region of the oncoprotein. Phenolic compounds and CagA exhibited significant affinity energy (-7.9 to -9.1 kcal/mol): CagA/kaempferol formed 28 chemical bonds, CagA/myricetin formed 18 chemical bonds, CagA/quercetin formed 16 chemical bonds, CagA/ponciretin formed 13 chemical bonds, and CagA/chlorogenic acid formed 17 chemical bonds. Although none of the phenolic compounds directly bound to the amino acid residues of the K-Xn-R-X-R membrane binding motif, all of them bound to residues, mostly positively or negatively charged, located near this region. CONCLUSION In silico, the tested phenolic compounds formed stable complexes with CagA. Therefore, they could be tested in vitro and/or in vivo to validate the findings, and to assess interference in CagA/cellular target interactions and in the oncogenic differentiation of gastric cells.
Collapse
Affiliation(s)
| | | | - Fabrício Freire de Melo
- Universidade Federal da Bahia, Instituto Multidisciplinar em Saúde-Campus Anísio Teixeira, Vitória da Conquista 45029-094, Brazil
| | - Ana Carla Zarpelon-Schutz
- Universidade Federal do Paraná, Campus Toledo, Toledo 85919-899, Brazil
- Universidade Federal do Paraná-Setor Palotina, Programa de Pós-graduação em Biotecnologia, Palotina 85950-000, Brazil
| | - Kádima Nayara Teixeira
- Universidade Federal do Paraná, Campus Toledo, Toledo 85919-899, Brazil
- Universidade Federal do Paraná-Setor Palotina, Programa de Pós-graduação em Biotecnologia, Palotina 85950-000, Brazil
| |
Collapse
|
13
|
Kostelecka K, Bryliński Ł, Komar O, Michalczyk J, Miłosz A, Biłogras J, Woliński F, Forma A, Baj J. An Overview of the Spices Used for the Prevention and Potential Treatment of Gastric Cancer. Cancers (Basel) 2024; 16:1611. [PMID: 38672692 PMCID: PMC11049028 DOI: 10.3390/cancers16081611] [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: 03/10/2024] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024] Open
Abstract
Gastric cancer (GC) ranks third in terms of cancer-related deaths and is the fifth most commonly diagnosed type of cancer. Its risk factors include Helicobacter pylori infection, Epstein-Barr virus infection, the consumption of broiled and charbroiled animal meats, salt-preserved and smoke-enhanced foods, alcohol drinking, tobacco smoking, exposure to ionizing radiation, and positive family history. The limited effectiveness of conventional therapies and the widespread risk factors of GC encourage the search for new methods of treatment and prevention. In the quest for cheap and commonly available medications, numerous studies focus on herbal medicine, traditional brews, and spices. In this review, we outline the potential use of spices, including turmeric, ginger, garlic, black cumin, chili pepper, saffron, black pepper, rosemary, galangal, coriander, wasabi, cinnamon, oregano, cardamom, fenugreek, caraway, clove, dill, thyme, Piper sarmentosum, basil, as well as the compounds they contain, in the prevention and treatment of GC. We present the potential molecular mechanisms responsible for the effectivity of a given seasoning substance and their impact on GC cells. We discuss their potential effects on proliferation, apoptosis, and migration. For most of the spices discussed, we also outline the unavailability and side effects of their use.
Collapse
Affiliation(s)
- Katarzyna Kostelecka
- Department of Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland; (K.K.); (Ł.B.); (O.K.); (J.M.); (A.M.); (J.B.); (J.B.)
| | - Łukasz Bryliński
- Department of Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland; (K.K.); (Ł.B.); (O.K.); (J.M.); (A.M.); (J.B.); (J.B.)
| | - Olga Komar
- Department of Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland; (K.K.); (Ł.B.); (O.K.); (J.M.); (A.M.); (J.B.); (J.B.)
| | - Justyna Michalczyk
- Department of Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland; (K.K.); (Ł.B.); (O.K.); (J.M.); (A.M.); (J.B.); (J.B.)
| | - Agata Miłosz
- Department of Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland; (K.K.); (Ł.B.); (O.K.); (J.M.); (A.M.); (J.B.); (J.B.)
| | - Jan Biłogras
- Department of Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland; (K.K.); (Ł.B.); (O.K.); (J.M.); (A.M.); (J.B.); (J.B.)
| | - Filip Woliński
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Alicja Forma
- Department of Forensic Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland;
| | - Jacek Baj
- Department of Anatomy, Medical University of Lublin, ul. Jaczewskiego 4, 20-090 Lublin, Poland; (K.K.); (Ł.B.); (O.K.); (J.M.); (A.M.); (J.B.); (J.B.)
| |
Collapse
|
14
|
Huang J, Chen J, Li J. Quercetin promotes ATG5-mediating autophagy-dependent ferroptosis in gastric cancer. J Mol Histol 2024; 55:211-225. [PMID: 38441713 DOI: 10.1007/s10735-024-10186-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 02/26/2024] [Indexed: 04/05/2024]
Abstract
Quercetin has been documented to possess a multitude of pharmacological effects, encompassing antioxidant, antiviral, antimicrobial, and anti-inflammatory properties. Nevertheless, the exact molecular mechanisms responsible for the anti-tumor properties of quercetin remain to be fully explicated. To this end, quercetin was administered to gastric cancer cells (in vitro) AGS and MKN45, as well as BALB/c mice (in vivo). The proliferation ability of cells was evaluated using cholecystokinin octapeptide (CCK-8) and colony formation assays. The evaluation of ferroptosis involved the measurement of iron, malondialdehyde (MDA), and lipid reactive oxygen species. Autophagy and apoptosis were evaluated using immunofluorescence staining, western blotting, and flow cytometry analysis. Our findings indicate that quercetin significantly inhibited cell viability and tumor volume compared to the control group. Additionally, quercetin was found to decrease glutathione (GSH), malondialdehyde, and reactive oxygen species (ROS) levels while suppressing beclin1 and LC3B levels in cancer cells. Remarkably, the utilization of siATG5 was found to reverse all the aforementioned effects of quercetin. Ultimately, the effects of quercetin on gastric cancer were validated. In summary, our findings provide evidence that quercetin facilitates autophagy-mediated ferroptosis in gastric cancer.
Collapse
Affiliation(s)
- Ju Huang
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Shuai Fu Community, Dong Cheng District, Beijing, 100730, China
| | - Jian Chen
- Department of Oncology, the Affiliated Yantai Yuhuangding Hospital of Qingdao University, No.20, Yuhuangding East Road, Zhifu District, Yantai, 264000, Shandong, China.
| | - Jingnan Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences Peking Union Medical College, Shuai Fu Community, Dong Cheng District, Beijing, 100730, China.
| |
Collapse
|
15
|
Cao X, Cheng XW, Liu YY, Dai HW, Gan RY. Inhibition of pathogenic microbes in oral infectious diseases by natural products: Sources, mechanisms, and challenges. Microbiol Res 2024; 279:127548. [PMID: 38016378 DOI: 10.1016/j.micres.2023.127548] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 10/31/2023] [Accepted: 11/10/2023] [Indexed: 11/30/2023]
Abstract
The maintenance of oral health is of utmost importance for an individual's holistic well-being and standard of living. Within the oral cavity, symbiotic microorganisms actively safeguard themselves against potential foreign diseases by upholding a multifaceted equilibrium. Nevertheless, the occurrence of an imbalance can give rise to a range of oral infectious ailments, such as dental caries, periodontitis, and oral candidiasis. Presently, clinical interventions encompass the physical elimination of pathogens and the administration of antibiotics to regulate bacterial and fungal infections. Given the limitations of various antimicrobial drugs frequently employed in dental practice, the rising incidence of oral inflammation, and the escalating bacterial resistance to antibiotics, it is imperative to explore alternative remedies that are dependable, efficacious, and affordable for the prevention and management of oral infectious ailments. There is an increasing interest in the creation of novel antimicrobial agents derived from natural sources, which possess attributes such as safety, cost-effectiveness, and minimal adverse effects. This review provides a comprehensive overview of the impact of natural products on the development and progression of oral infectious diseases. Specifically, these products exert their influences by mitigating dental biofilm formation, impeding the proliferation of oral pathogens, and hindering bacterial adhesion to tooth surfaces. The review also encompasses an examination of the various classes of natural products, their antimicrobial mechanisms, and their potential therapeutic applications and limitations in the context of oral infections. The insights garnered from this review can support the promising application of natural products as viable therapeutic options for managing oral infectious diseases.
Collapse
Affiliation(s)
- Xin Cao
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China
| | - Xing-Wang Cheng
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yin-Ying Liu
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A⁎STAR), 31 Biopolis Way, Singapore 138669, Singapore; Department of Food Science and Technology, Faculty of Science, National University of Singapore, 2 Science Drive 2, Singapore 117542, Singapore
| | - Hong-Wei Dai
- College of Stomatology, Chongqing Medical University, Chongqing 401147, China; Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Chongqing 401147, China; Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing 401147, China.
| | - Ren-You Gan
- Singapore Institute of Food and Biotechnology Innovation (SIFBI), Agency for Science, Technology and Research (A⁎STAR), 31 Biopolis Way, Singapore 138669, Singapore; Department of Food Science and Technology, Faculty of Science, National University of Singapore, 2 Science Drive 2, Singapore 117542, Singapore.
| |
Collapse
|
16
|
Liu M, Gao H, Miao J, Zhang Z, Zheng L, Li F, Zhou S, Zhang Z, Li S, Liu H, Sun J. Helicobacter pylori infection in humans and phytotherapy, probiotics, and emerging therapeutic interventions: a review. Front Microbiol 2024; 14:1330029. [PMID: 38268702 PMCID: PMC10806011 DOI: 10.3389/fmicb.2023.1330029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024] Open
Abstract
The global prevalence of Helicobacter pylori (H. pylori) infection remains high, indicating a persistent presence of this pathogenic bacterium capable of infecting humans. This review summarizes the population demographics, transmission routes, as well as conventional and novel therapeutic approaches for H. pylori infection. The prevalence of H. pylori infection exceeds 30% in numerous countries worldwide and can be transmitted through interpersonal and zoonotic routes. Cytotoxin-related gene A (CagA) and vacuolar cytotoxin A (VacA) are the main virulence factors of H. pylori, contributing to its steep global infection rate. Preventative measures should be taken from people's living habits and dietary factors to reduce H. pylori infection. Phytotherapy, probiotics therapies and some emerging therapies have emerged as alternative treatments for H. pylori infection, addressing the issue of elevated antibiotic resistance rates. Plant extracts primarily target urease activity and adhesion activity to treat H. pylori, while probiotics prevent H. pylori infection through both immune and non-immune pathways. In the future, the primary research focus will be on combining multiple treatment methods to effectively eradicate H. pylori infection.
Collapse
Affiliation(s)
- Mengkai Liu
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Hui Gao
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Jinlai Miao
- First Institute of Oceanography Ministry of Natural Resources, Qingdao, China
| | - Ziyan Zhang
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Lili Zheng
- National Engineering Research Centre for Intelligent Electrical Vehicle Power System (Qingdao), College of Mechanical and Electronic Engineering, Qingdao University, Qingdao, China
| | - Fei Li
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Sen Zhou
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Zhiran Zhang
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Shengxin Li
- College of Life Sciences, Qingdao University, Qingdao, China
| | - He Liu
- College of Life Sciences, Qingdao University, Qingdao, China
| | - Jie Sun
- College of Life Sciences, Qingdao University, Qingdao, China
| |
Collapse
|
17
|
Jalali Z, Nejad Ebrahimi S, Rezadoost H. Identifying natural products for gastric cancer treatment through pharmacophore creation, 3D QSAR, virtual screening, and molecular dynamics studies. Daru 2023; 31:243-258. [PMID: 37733194 PMCID: PMC10624797 DOI: 10.1007/s40199-023-00480-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/06/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Gastric cancer (GC) is known as the fourth leading cause of cancer-related death and the fifth major cancer in the world, and this is a serious threat to general health all over the world. The lack of early detection markers results in a belated diagnosis, i.e. the final stages, which could be associated with the ineffectiveness of the treatment strategies, and naturally, it leads to poor prognosis. Even though a variety of treatments have been developed, there is a trend of studying traditional medicinal plants, due to the worrying side effect of drugs available in the market. METHODS In this study, pharmacophore generation and 3D-QSAR model were created using 50 compounds with anti-gastric cancer activity (with IC50 had been reported in the previous studies). RESULTS Based on three of the best pharmacophoric hypotheses, virtual screening was performed to discover the top anti-gastric cancer compounds from a database of 183,885 compounds. The selected compounds were used for molecular docking with three protein receptors 7BKG, 4F5B, and 4ZT1 to investigate the intermolecular interactions between these ligands and receptors. Finally, 21 lead compounds with the highest amount of docking score ranging from - 13.366 to -6.404 kcal/mol were selected, and then the ADME/Tox properties of these compounds were calculated. All these compounds have a fitness score above 1.8, a molecular weight of less than 500 g/mol, hydrogen bond donors up to 3, hydrogen bond acceptors up to 8.50, and logP of 1.013 to 4.174. Finally, molecular dynamic simulations for top-scoring ligand-receptor complexes were investigated. CONCLUSION These selected lead compounds have the most anti-gastric cancer effects among the 183,885 compounds in the database. Therefore, lead compounds might be considered for gastric cancer therapy in future studies.
Collapse
Affiliation(s)
- Zeinab Jalali
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Evin, 1983963113, Tehran, Iran
| | - Samad Nejad Ebrahimi
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Evin, 1983963113, Tehran, Iran.
| | - Hassan Rezadoost
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Evin, 1983963113, Tehran, Iran
| |
Collapse
|
18
|
Liu Z, Huang H, Yu Y, Li L, Shi X, Wang F. Exploring the mechanism of ellagic acid against gastric cancer based on bioinformatics analysis and network pharmacology. J Cell Mol Med 2023; 27:3878-3896. [PMID: 37794689 PMCID: PMC10718161 DOI: 10.1111/jcmm.17967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/17/2023] [Accepted: 08/30/2023] [Indexed: 10/06/2023] Open
Abstract
Ellagic acid (EA) is a natural polyphenolic compound. Recent studies have shown that EA has potential anticancer properties against gastric cancer (GC). This study aims to reveal the potential targets and mechanisms of EA against GC. This study adopted methods of bioinformatics analysis and network pharmacology, including the weighted gene co-expression network analysis (WGCNA), construction of protein-protein interaction (PPI) network, receiver operating characteristic (ROC) and Kaplan-Meier (KM) survival curve analysis, Gene Ontology (GO) function and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, molecular docking and molecular dynamics simulations (MDS). A total of 540 EA targets were obtained. Through WGCNA, we obtained a total of 2914 GC clinical module genes, combined with the disease database for screening, a total of 606 GC-related targets and 79 intersection targets of EA and GC were obtained by constructing Venn diagram. PPI network was constructed to identify 14 core candidate targets; TP53, JUN, CASP3, HSP90AA1, VEGFA, HRAS, CDH1, MAPK3, CDKN1A, SRC, CYCS, BCL2L1 and CDK4 were identified as the key targets of EA regulation of GC by ROC and KM curve analysis. The enrichment analysis of GO and KEGG pathways of key targets was performed, and they were mainly enriched in p53 signalling pathway, PI3K-Akt signalling pathway. The results of molecular docking and MDS showed that EA could effectively bind to 13 key targets to form stable protein-ligand complexes. This study revealed the key targets and molecular mechanisms of EA against GC and provided a theoretical basis for further study of the pharmacological mechanism of EA against GC.
Collapse
Affiliation(s)
- Zhiyao Liu
- Department of Rehabilitation MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Hailiang Huang
- Department of Rehabilitation MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Ying Yu
- Innovative Institute of Chinese Medicine and PharmacyShandong University of Traditional Chinese MedicineJinanChina
| | - Lingling Li
- Department of Rehabilitation MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Xin Shi
- Department of Rehabilitation MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Fangqi Wang
- Department of Rehabilitation MedicineShandong University of Traditional Chinese MedicineJinanChina
| |
Collapse
|
19
|
Akash S, Bayıl I, Mahmood S, Mukerjee N, Mili TA, Dhama K, Rahman MA, Maitra S, Mohany M, Al-Rejaie SS, Ali N, Semwal P, Sharma R. Mechanistic inhibition of gastric cancer-associated bacteria Helicobacter pylori by selected phytocompounds: A new cutting-edge computational approach. Heliyon 2023; 9:e20670. [PMID: 37876433 PMCID: PMC10590806 DOI: 10.1016/j.heliyon.2023.e20670] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 09/09/2023] [Accepted: 10/04/2023] [Indexed: 10/26/2023] Open
Abstract
Background Helicobacter pylori (H. pylori) is a persistent bacterial inhabitant in the stomachs of approximately half the global populace. This bacterium is directly linked to chronic gastritis, leading to a heightened risk of duodenal and gastric ulcer diseases, and is the predominant risk factor for gastric cancer - the second most common cause of cancer-related deaths globally. The increasing prevalence of antibiotic resistance necessitates the exploration of innovative treatment alternatives to mitigate the H. pylori menace. Methods Initiating our study, we curated a list of thirty phytochemicals based on previous literature and subjected them to molecular docking studies. Subsequently, eight phytocompounds-Glabridin, Isoliquiritin, Sanguinarine, Liquiritin, Glycyrrhetic acid, Beta-carotin, Diosgenin, and Sarsasapogenin-were meticulously chosen based on superior binding scores. These were further subjected to an extensive computational analysis encompassing ADMET profiling, drug-likeness evaluation, principal component analysis (PCA), and molecular dynamic simulations (MDs) in comparison with the conventional drug, Mitomycin. Results The natural compounds investigated demonstrated superior docking affinities to H. pylori targets compared to the standard Mitomycin. Notably, the phytocompounds Diosgenin and Sarsasapogenin stood out due to their exceptional binding affinities and pharmacokinetic properties, including favorable ADMET profiles. Conclusion Our comprehensive and technologically-advanced approach showcases the potential of identified phytocompounds as pioneering therapeutic agents against H. pylori-induced gastric malignancies. In light of our promising in silico results, we recommend these natural compounds as potential candidates for advancing H. pylori-targeted drug development. Given their potential, we strongly advocate for subsequent in vitro and in vivo studies to validate their therapeutic efficacy against this formidable gastrointestinal bacterium.
Collapse
Affiliation(s)
- Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Birulia, 1216, Ashulia, Dhaka, Bangladesh
| | - Imren Bayıl
- Department of Bioinformatics and Computational Biology, Gaziantep University, Turkey
| | - Sajjat Mahmood
- Department of Microbiology, Jagannath University, Chittaranjan Avenue in Sadarghat, Dhaka, 1100, Bangladesh
| | - Nobendu Mukerjee
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute Of Medical and Technical Sciences, Chennai, India
- Department of Microbiology, West Bengal State University, West Bengal, Kolkata, 700126, India
- Department of Health Sciences, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Tamanna Akter Mili
- Department of Pharmacy, University of Asia Pacific, 74/A Green Rd, Dhaka, 1205, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute (IVRI), Izatnagar, 243122, Bareilly, Uttar Pradesh, India
| | | | - Swastika Maitra
- Department of Microbiology, Adamas University, West Bengal, Kolkata, 700126, India
| | - Mohamed Mohany
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh, 1145, Saudi Arabia
| | - Salim S. Al-Rejaie
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh, 1145, Saudi Arabia
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 55760, Riyadh, 1145, Saudi Arabia
| | - Prabhakar Semwal
- Department of Biotechnology, Graphic Era University, Dehradun, Uttarakhand, 248002, India
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Science, Banaras Hindu University, Varanasi, 221005, India
| |
Collapse
|
20
|
Gupta A, Shetty S, Mutalik S, Chandrashekar H R, K N, Mathew EM, Jha A, Mishra B, Rajpurohit S, Ravi G, Saha M, Moorkoth S. Treatment of H. pylori infection and gastric ulcer: Need for novel Pharmaceutical formulation. Heliyon 2023; 9:e20406. [PMID: 37810864 PMCID: PMC10550623 DOI: 10.1016/j.heliyon.2023.e20406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 09/21/2023] [Accepted: 09/22/2023] [Indexed: 10/10/2023] Open
Abstract
Peptic ulcer disease (PUD) is one of the most prevalent gastro intestinal disorder which often leads to painful sores in the stomach lining and intestinal bleeding. Untreated Helicobacter pylori (H. pylori) infection is one of the major reasons for chronic PUD which, if left untreated, may also result in gastric cancer. Treatment of H. pylori is always a challenge to the treating doctor because of the poor bioavailability of the drug at the inner layers of gastric mucosa where the bacteria resides. This results in ineffective therapy and antibiotic resistance. Current treatment regimens available for gastric ulcer and H. pylori infection uses a combination of multiple antimicrobial agents, proton pump inhibitors (PPIs), H2-receptor antagonists, dual therapy, triple therapy, quadruple therapy and sequential therapy. This polypharmacy approach leads to patient noncompliance during long term therapy. Management of H. pylori induced gastric ulcer is a burning issue that necessitates alternative treatment options. Novel formulation strategies such as extended-release gastro retentive drug delivery systems (GRDDS) and nanoformulations have the potential to overcome the current bioavailability challenges. This review discusses the current status of H. pylori treatment, their limitations and the formulation strategies to overcome these shortcomings. Authors propose here an innovative strategy to improve the H. pylori eradication efficiency.
Collapse
Affiliation(s)
- Ashutosh Gupta
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Shiran Shetty
- Department of Gastroenterology and Hepatology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Srinivas Mutalik
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Raghu Chandrashekar H
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Nandakumar K
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Elizabeth Mary Mathew
- School of Pharmacy, Faculty of Health Sciences, University of Botswana, Gaborone, Botswana
| | - Abhishek Jha
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Brahmeshwar Mishra
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, Uttar Pradesh, India
| | - Siddheesh Rajpurohit
- Department of Gastroenterology and Hepatology, Kasturba Medical College, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Gundawar Ravi
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Moumita Saha
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Sudheer Moorkoth
- Department of Pharmaceutical Quality Assurance, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
21
|
Carradori S, Ammazzalorso A, Niccolai S, Tanini D, D’Agostino I, Melfi F, Capperucci A, Grande R, Sisto F. Nature-Inspired Compounds: Synthesis and Antibacterial Susceptibility Testing of Eugenol Derivatives against H. pylori Strains. Pharmaceuticals (Basel) 2023; 16:1317. [PMID: 37765124 PMCID: PMC10534785 DOI: 10.3390/ph16091317] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/10/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
The antimicrobial properties of one of the most important secondary metabolites, Eugenol (EU), inspired us to design and synthesize three different series of derivatives enhancing its parent compound's anti-Helicobacter pylori activity. Thus, we prepared semisynthetic derivatives through (A) diazo aryl functionalization, (B) derivatization of the hydroxy group of EU, and (C) elongation of the allyl radical by incorporating a chalcogen atom. The antibacterial evaluation was performed on the reference NCTC 11637 strain and on three drug-resistant clinical isolates and the minimal inhibitory and bactericidal concentrations (MICs and MBCs) highlight the role of chalcogens in enhancing the antimicrobial activity (less than 4 µg/mL for some compounds) of the EU scaffold (32-64 µg/mL).
Collapse
Affiliation(s)
- Simone Carradori
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (S.C.); (F.M.); (R.G.)
| | - Alessandra Ammazzalorso
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (S.C.); (F.M.); (R.G.)
| | - Sofia Niccolai
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3–13, 50019 Sesto Fiorentino, Italy; (S.N.); (D.T.); (A.C.)
| | - Damiano Tanini
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3–13, 50019 Sesto Fiorentino, Italy; (S.N.); (D.T.); (A.C.)
| | - Ilaria D’Agostino
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy;
| | - Francesco Melfi
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (S.C.); (F.M.); (R.G.)
| | - Antonella Capperucci
- Department of Chemistry “Ugo Schiff”, University of Florence, Via della Lastruccia 3–13, 50019 Sesto Fiorentino, Italy; (S.N.); (D.T.); (A.C.)
| | - Rossella Grande
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (S.C.); (F.M.); (R.G.)
| | - Francesca Sisto
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, Via Pascal 36, 20133 Milan, Italy;
| |
Collapse
|
22
|
Frański R, Beszterda-Buszczak M. Comment on Villalva et al. Antioxidant, Anti-Inflammatory, and Antibacterial Properties of an Achillea millefolium L. Extract and Its Fractions Obtained by Supercritical Anti-Solvent Fractionation against Helicobacter pylori. Antioxidants 2022, 11, 1849. Antioxidants (Basel) 2023; 12:1226. [PMID: 37371956 DOI: 10.3390/antiox12061226] [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: 11/08/2022] [Revised: 04/21/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
Abstract
Villalva et al. evaluated the potential utility of an Achillea millefolium (yarrow) extract in the control of H. pylori infection. The agar-well diffusions bioassay was applied to determine the antimicrobial activity of yarrow extracts. The supercritical anti-solvent fractionation process of yarrow extract was made to give two different fractions with polar phenolic compounds and monoterpenes and sesquiterpenes, respectively. Phenolic compounds were identified by HPLC-ESIMS by using the accurate masses of [M-H]- ions and the characteristic product ions. However, some of the reported product ions seem to be disputable, as described below.
Collapse
Affiliation(s)
- Rafał Frański
- Faculty of Chemistry, Adam Mickiewicz University, Uniwersytetu Poznańskiego 8, 61-614 Poznań, Poland
| | - Monika Beszterda-Buszczak
- Department of Food Biochemistry and Analysis, Poznań University of Life Sciences, Mazowiecka 48, 60-623 Poznań, Poland
| |
Collapse
|
23
|
Dinat S, Orchard A, Van Vuuren S. A scoping review of African natural products against gastric ulcers and Helicobacter pylori. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115698. [PMID: 36174808 DOI: 10.1016/j.jep.2022.115698] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/25/2022] [Accepted: 09/02/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE African traditional medicine is one of the oldest and most diverse practices for treating ailments and numerous natural products have been recommended for gastric ulcer treatment. Helicobacter pylori is the main causative organism implicated in several diseases, most notably in causing inflammation and the onset of gastric ulcers. Current H. pylori treatment methods are losing efficacy as H. pylori rapidly gains resistance to antibiotics. Hence, a search into natural products and their historical traditional efficacy for the treatment of gastric ulcers is of interest. AIM OF THE STUDY This review aimed to summarise the African use of natural products, including medicinal plants noted in ethnobotanical reviews, used traditionally to treat gastric ulcers, and highlights the investigations into the anti-H. pylori activity of medicinal plants and bee products found in Africa. METHODOLOGY A systematic review was carried out to identify natural products, including those used traditionally in Africa to treat gastric ulcers, and to correlate this with scientific investigations into the anti-H. pylori activity of natural products used in Africa. RESULTS A total of 107 literature sources describing the traditional use of medicinal plants in gastric ulcer treatment were found, from which 360 medicinal plants were identified. Of the plants used traditionally for gastric ulcer treatment, 11% were investigated either in vitro or in vivo for anti-ulcer and anti-H. pylori activity. Of the 122 medicinal plants eliciting antimicrobial or anti-ulcer activity, Hibiscus sabdariffa L. calyx extract and Terminalia macroptera Guill. & Perr. root extract were found to have the most noteworthy antimicrobial activity, with minimum inhibitory concentrations (MICs) of 0.01 mg/mL and 0.03 mg/mL respectively. The essential oils of Piper longum L. and Pachira aquatica Aubl. displayed the most notable in vitro anti-H. pylori activity (MIC of 0.01 mg/mL and 0.02 mg/mL). Several in vivo studies found medicinal plant extracts effective in reducing the H. pylori load along the gastric mucosa. The South African honey variants, Pure Honey and Champagne Royal Train (common names given by supplier) were the most antimicrobially effective (MIC of 0.01-10.0%, 0.63-10.00% v/v) in inhibiting H. pylori when assessed in vitro. CONCLUSION These results highlight the potential of natural products to inhibit H. pylori growth and serve as a possible stepping-stone in understanding the management of ulcers. Furthermore, effective natural product treatment or prophylactic use for preventing H. pylori growth may provide a more affordable option for African populations.
Collapse
Affiliation(s)
- S Dinat
- Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193, South Africa
| | - A Orchard
- Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193, South Africa
| | - S Van Vuuren
- Department of Pharmacy and Pharmacology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193, South Africa.
| |
Collapse
|
24
|
Editorial: Special issue: Advances in microbial pathogenesis. Microb Pathog 2023; 174:105926. [PMID: 36464076 DOI: 10.1016/j.micpath.2022.105926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
25
|
Moreira H, Dobosz A, Cwynar-Zając Ł, Nowak P, Czyżewski M, Barg M, Reichert P, Królikowska A, Barg E. Unraveling the role of Breg cells in digestive tract cancer and infectious immunity. Front Immunol 2022; 13:981847. [PMID: 36618354 PMCID: PMC9816437 DOI: 10.3389/fimmu.2022.981847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 12/02/2022] [Indexed: 12/25/2022] Open
Abstract
Over the past two decades, regulatory B cells (Breg cells or Bregs) have emerged as an immunosuppressive subset of B lymphocytes playing a key role in inflammation, infection, allergy, transplantation, and cancer. However, the involvement of Bregs in various pathological conditions of the gastrointestinal tract is not fully understood and is the subject of much recent research. In this review, we aimed to summarize the current state of knowledge about the origin, phenotype, and suppressive mechanisms of Bregs. The relationship between the host gut microbiota and the function of Bregs in the context of the disturbance of mucosal immune homeostasis is also discussed. Moreover, we focused our attention on the role of Bregs in certain diseases and pathological conditions related to the digestive tract, especially Helicobacter pylori infection, parasitic diseases (leishmaniasis and schistosomiasis), and gastrointestinal neoplasms. Increasing evidence points to a relationship between the presence and number of Bregs and the severity and progression of these pathologies. As the number of cases is increasing year by year, also among young people, it is extremely important to understand the role of these cells in the digestive tract.
Collapse
Affiliation(s)
- Helena Moreira
- Department of Medical Sciences Foundation, Wroclaw Medical University, Wroclaw, Poland,*Correspondence: Helena Moreira, ; Agnieszka Dobosz,
| | - Agnieszka Dobosz
- Department of Medical Sciences Foundation, Wroclaw Medical University, Wroclaw, Poland,*Correspondence: Helena Moreira, ; Agnieszka Dobosz,
| | - Łucja Cwynar-Zając
- Department of Medical Sciences Foundation, Wroclaw Medical University, Wroclaw, Poland
| | - Paulina Nowak
- Faculty of Pharmacy, Wroclaw Medical University, Wroclaw, Poland
| | - Marek Czyżewski
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Marta Barg
- Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Paweł Reichert
- Department of Trauma Surgery, Clinical Department of Trauma and Hand Surgery, Faculty of Medicine, Wroclaw Medical University, Wroclaw, Poland
| | - Aleksandra Królikowska
- Ergonomics and Biomedical Monitoring Laboratory, Department of Physiotherapy, Faculty of Health Sciences, Wroclaw Medical University, Wroclaw, Poland
| | - Ewa Barg
- Department of Medical Sciences Foundation, Wroclaw Medical University, Wroclaw, Poland
| |
Collapse
|
26
|
Preparation, Characterization, and Anti-Adhesive Activity of Sulfate Polysaccharide from Caulerpa lentillifera against Helicobacter pylori. Polymers (Basel) 2022; 14:polym14224993. [PMID: 36433125 PMCID: PMC9697858 DOI: 10.3390/polym14224993] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/11/2022] [Accepted: 11/12/2022] [Indexed: 11/19/2022] Open
Abstract
In the gastric mucosa, chronic inflammation due to Helicobacter pylori infection promotes gastrocarcinogenesis. Polysaccharides of Caulerpa lentillifera are well-characterized by broad antimicrobial activity and anti-inflammatory potentials. The present study was undertaken to investigate whether the low molecular sulfate polysaccharides of C. lentillifera (CLCP) exhibit any anti-adhesive activity against H. pylori. After a hot water extraction and purification process, two purified polysaccharide fractions (CLCP-1 and CLCP2) were studied based on structural characterization and bioactivity determination. The results implied that except for the molar ratio, CLCP-1 and CLCP-2 contain high sulfate, mannose, galactose, xylose, glucose levels, and low protein levels. The molecular weight and Fourier transform infrared spectroscopy (FT-IR) assays confirmed that CLCP-1 and CLCP-2 are sulfate polysaccharides with an average molecular weight (Mw) of 963.15 and 648.42 kDa, respectively. In addition, CLCP-1 and CLCP-2 exhibited stronger antibacterial activity against H. pylori. CLCP-1 and CLCP-2 could significantly promote macrophage proliferation and decrease the production of nitric oxide (NO) through downregulated expression of inducible nitric oxide synthase (iNOS). Meanwhile, CLCP-1 and CLCP-2 in this study showed efficiently protected gastric adenocarcinoma (AGS) cells against H. pylori with the inhibition of the IL-8/NF-κB axis. These findings suggested the effect of Caulerpa lentillifera polysaccharides on H. pylori adhesion, a potential supply of nutrients for eradication therapy through the reduction of cell count and inflammation.
Collapse
|