1
|
Purdel C, Margină D, Adam-Dima I, Ungurianu A. The Beneficial Effects of Dietary Interventions on Gut Microbiota-An Up-to-Date Critical Review and Future Perspectives. Nutrients 2023; 15:5005. [PMID: 38068863 PMCID: PMC10708505 DOI: 10.3390/nu15235005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/22/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Different dietary interventions, especially intermittent fasting, are widely used and promoted by physicians; these regimens have been studied lately for their impact on the gut microbiota composition/function and, consequently, on the general physiopathological processes of the host. Studies are showing that dietary components modulate the microbiota, and, at the same time, the host metabolism is deeply influenced by the different products resulting from nutrient transformation in the microbiota compartment. This reciprocal relationship can potentially influence even drug metabolism for chronic drug regimens, significantly impacting human health/disease. Recently, the influence of various dietary restrictions on the gut microbiota and the differences between the effects were investigated. In this review, we explored the current knowledge of different dietary restrictions on animal and human gut microbiota and the impact of these changes on human health.
Collapse
Affiliation(s)
- Carmen Purdel
- Department of Toxicology, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (C.P.); (I.A.-D.)
| | - Denisa Margină
- Department of Biochemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania;
| | - Ines Adam-Dima
- Department of Toxicology, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (C.P.); (I.A.-D.)
| | - Anca Ungurianu
- Department of Biochemistry, Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania;
| |
Collapse
|
2
|
Zhang Y, Zeng M, Zhang X, Yu Q, Zeng W, Yu B, Gan J, Zhang S, Jiang X. Does an apple a day keep away diseases? Evidence and mechanism of action. Food Sci Nutr 2023; 11:4926-4947. [PMID: 37701204 PMCID: PMC10494637 DOI: 10.1002/fsn3.3487] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 09/14/2023] Open
Abstract
Apples and their products exemplify the recently reemphasized link between dietary fruit intake and the alleviation of human disease. Their consumption does indeed improve human health due to their high phytochemical content. To identify potentially relevant articles from clinical trials, some epidemiological studies and meta-analyses, and in vitro and in vivo studies (cell cultures and animal models), PubMed was searched from January 1, 2012, to May 15, 2022. This review summarized the potential effects of apple and apple products (juices, puree, pomace, dried apples, extracts rich in apple bioactives and single apple bioactives) on health. Apples and apple products have protective effects against cardiovascular diseases, cancer, as well as mild cognitive impairment and promote hair growth, healing of burn wounds, improve the oral environment, prevent niacin-induced skin flushing, promote the relief of UV-induced skin pigmentation, and improve the symptoms of atopic dermatitis as well as cedar hay fever among others. These effects are associated with various mechanisms, such as vascular endothelial protection, blood lipids lowering, anti-inflammatory, antioxidant, antiapoptotic, anti-invasion, and antimetastatic effects. Meanwhile, it has provided an important reference for the application and development of medicine, nutrition, and other fields.
Collapse
Affiliation(s)
- Yue Zhang
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Miao Zeng
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Xiaolu Zhang
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Qun Yu
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Wenyun Zeng
- Department of PathologyTianjin Union Medical CenterTianjinChina
| | - Bin Yu
- School of International EducationTianjin University of Chinese MedicineTianjinChina
| | - Jiali Gan
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Shiwu Zhang
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
- Department of PathologyTianjin Union Medical CenterTianjinChina
| | - Xijuan Jiang
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| |
Collapse
|
3
|
Messina M, Iacumin L, Pascon G, Tulli F, Tibaldi E, Cardinaletti G. Effect of feed restriction and refeeding on body condition, digestive functionality and intestinal microbiota in rainbow trout (Oncorhynchus mykiss). FISH PHYSIOLOGY AND BIOCHEMISTRY 2023; 49:169-189. [PMID: 36680627 PMCID: PMC9935662 DOI: 10.1007/s10695-023-01170-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 01/05/2023] [Indexed: 06/17/2023]
Abstract
The aim of the present work was to investigate the influence of fasting and refeeding on body condition, gut physiology and microbiota in reared O. mykiss. Ninety-six fish were randomly allotted among three groups subjected to different feeding plan: C (control, fed for 5 weeks); R (restricted ration over 3 weeks followed by 2 weeks feeding); F (fasted over 3 weeks followed by 2 weeks feeding) in a well's fresh water flow-through rearing plan. Sampling occurred at 0, 1, 2, 4, 7, 14 days during the refeeding period. At day 0 and throughout the feeding period until day 14, the weight of the fish was significantly affected by the feeding restriction. Feed deprivation reduced significantly the viscerosomatic and hepatosomatic indexes. Brush border membrane enzymes' specific activity was modulated by feeding regimes until day 7, to level in all experimental groups at day 14. At the end of the restricted/fasted period, the microbiota of the C group was made up of 70% of Actinobacteria, 24% of Proteobacteria, 4.2% of Firmicutes and < 1% of Bacteroides, while the restricted and fasted group were characterized by a strong reduction of Actinobacteria, and a significant increase in Bacteroidetes and Firmicutes. The feed deprivation determined a dysbiosis, allowing the development of different commensal or pathogenic bacteria. In conclusion, the effects of 2 weeks of feed deprivation, excluding those related to body weight, are gradually mitigated by refeeding, which allows the restoration of digestive functions and a healthy intestinal microbiota.
Collapse
Affiliation(s)
- Maria Messina
- Department of Agricultural, Food, Environmental and Animal Science, University of Udine, Udine, Italy
| | - Lucilla Iacumin
- Department of Agricultural, Food, Environmental and Animal Science, University of Udine, Udine, Italy
| | - Giulia Pascon
- Department of Agricultural, Food, Environmental and Animal Science, University of Udine, Udine, Italy
| | - Francesca Tulli
- Department of Agricultural, Food, Environmental and Animal Science, University of Udine, Udine, Italy
| | - Emilio Tibaldi
- Department of Agricultural, Food, Environmental and Animal Science, University of Udine, Udine, Italy
| | - Gloriana Cardinaletti
- Department of Agricultural, Food, Environmental and Animal Science, University of Udine, Udine, Italy
| |
Collapse
|
4
|
Ismail S, Alsowayeh N, Abbasi HW, Albutti A, Tahir ul Qamar M, Ahmad S, Raza RZ, Sadia K, Abbasi SW. Pan-Genome-Assisted Computational Design of a Multi-Epitopes-Based Vaccine Candidate against Helicobacter cinaedi. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:11579. [PMID: 36141842 PMCID: PMC9517149 DOI: 10.3390/ijerph191811579] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/18/2022] [Accepted: 08/30/2022] [Indexed: 06/16/2023]
Abstract
Helicobacter cinaedi is a Gram-negative bacterium from the family Helicobacteraceae and genus Helicobacter. The pathogen is a causative agent of gastroenteritis, cellulitis, and bacteremia. The increasing antibiotic resistance pattern of the pathogen prompts the efforts to develop a vaccine to prevent dissemination of the bacteria and stop the spread of antibiotic resistance (AR) determinants. Herein, a pan-genome analysis of the pathogen strains was performed to shed light on its core genome and its exploration for potential vaccine targets. In total, four vaccine candidates (TonB dependent receptor, flagellar hook protein FlgE, Hcp family type VI secretion system effector, flagellar motor protein MotB) were identified as promising vaccine candidates and subsequently subjected to an epitopes' mapping phase. These vaccine candidates are part of the pathogen core genome: they are essential, localized at the pathogen surface, and are antigenic. Immunoinformatics was further applied on the selected vaccine proteins to predict potential antigenic, non-allergic, non-toxic, virulent, and DRB*0101 epitopes. The selected epitopes were then fused using linkers to structure a multi-epitopes' vaccine construct. Molecular docking simulations were conducted to determine a designed vaccine binding stability with TLR5 innate immune receptor. Further, binding free energy by MMGB/PBSA and WaterSwap was employed to examine atomic level interaction energies. The designed vaccine also stimulated strong humoral and cellular immune responses as well as interferon and cytokines' production. In a nutshell, the designed vaccine is promising in terms of immune responses' stimulation and could be an ideal candidate for experimental analysis due to favorable physicochemical properties.
Collapse
Affiliation(s)
- Saba Ismail
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Noorah Alsowayeh
- Department of Biology, College of Education (Majmaah), Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Hyder Wajid Abbasi
- Pakistan Institute of Medical Sciences, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad 44000, Pakistan
| | - Aqel Albutti
- Department of Medical Biotechnology, College of Applied Medical Sciences, Qassim University, Buraydah 51452, Saudi Arabia
| | - Muhammad Tahir ul Qamar
- Department of Bioinformatics and Biotechnology, Government College University Faisalabad, Faisalabad 38000, Pakistan
| | - Sajjad Ahmad
- Department of Health and Biological Sciences, Abasyn University, Peshawar 25000, Pakistan
| | - Rabail Zehra Raza
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| | - Khulah Sadia
- Department of Biosciences, COMSAT University, Islamabad 45550, Pakistan
| | - Sumra Wajid Abbasi
- Department of Biological Sciences, National University of Medical Sciences, Rawalpindi 46000, Pakistan
| |
Collapse
|
5
|
Malfertheiner P, Megraud F, Rokkas T, Gisbert JP, Liou JM, Schulz C, Gasbarrini A, Hunt RH, Leja M, O'Morain C, Rugge M, Suerbaum S, Tilg H, Sugano K, El-Omar EM. Management of Helicobacter pylori infection: the Maastricht VI/Florence consensus report. Gut 2022; 71:gutjnl-2022-327745. [PMID: 35944925 DOI: 10.1136/gutjnl-2022-327745] [Citation(s) in RCA: 594] [Impact Index Per Article: 198.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/21/2022] [Indexed: 01/06/2023]
Abstract
Helicobacter pyloriInfection is formally recognised as an infectious disease, an entity that is now included in the International Classification of Diseases 11th Revision. This in principle leads to the recommendation that all infected patients should receive treatment. In the context of the wide clinical spectrum associated with Helicobacter pylori gastritis, specific issues persist and require regular updates for optimised management.The identification of distinct clinical scenarios, proper testing and adoption of effective strategies for prevention of gastric cancer and other complications are addressed. H. pylori treatment is challenged by the continuously rising antibiotic resistance and demands for susceptibility testing with consideration of novel molecular technologies and careful selection of first line and rescue therapies. The role of H. pylori and antibiotic therapies and their impact on the gut microbiota are also considered.Progress made in the management of H. pylori infection is covered in the present sixth edition of the Maastricht/Florence 2021 Consensus Report, key aspects related to the clinical role of H. pylori infection were re-evaluated and updated. Forty-one experts from 29 countries representing a global community, examined the new data related to H. pylori infection in five working groups: (1) indications/associations, (2) diagnosis, (3) treatment, (4) prevention/gastric cancer and (5) H. pylori and the gut microbiota. The results of the individual working groups were presented for a final consensus voting that included all participants. Recommendations are provided on the basis of the best available evidence and relevance to the management of H. pylori infection in various clinical fields.
Collapse
Affiliation(s)
- Peter Malfertheiner
- Medical Department 2, LMU, Munchen, Germany
- Department of Radiology, LMU, Munchen, Germany
| | - Francis Megraud
- INSERM U853 UMR BaRITOn, University of Bordeaux, Bordeaux, France
| | - Theodore Rokkas
- Gastroenterology, Henry Dunant Hospital Center, Athens, Greece
- Medical School, European University, Nicosia, Cyprus
| | - Javier P Gisbert
- Gastroenterology, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IP), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - Jyh-Ming Liou
- Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Christian Schulz
- Medical Department 2, LMU, Munchen, Germany
- Partner Site Munich, DZIF, Braunschweig, Germany
| | - Antonio Gasbarrini
- Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Gemelli IRCCS, Università Cattolica del Sacro Cuore Facoltà di Medicina e Chirurgia, Roma, Italy
| | - Richard H Hunt
- Medicine, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, Hamilton, Ontario, Canada
| | - Marcis Leja
- Faculty of Medicine, University of Latvia, Riga, Latvia
- Institute of Clinical and Preventive Medicine, University of Latvia, Riga, Latvia
| | - Colm O'Morain
- Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Massimo Rugge
- Department of Medicine (DIMED), Surgical Pathology & Cytopathology Unit, University of Padova, Padova, Italy
- Veneto Tumor Registry (RTV), Padova, Italy
| | - Sebastian Suerbaum
- Partner Site Munich, DZIF, Braunschweig, Germany
- Max von Pettenkofer Institute, LMU, Munchen, Germany
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medizinische Universitat Innsbruck, Innsbruck, Austria
| | - Kentaro Sugano
- Department of Medicine, Jichi Medical School, Tochigi, Japan
| | - Emad M El-Omar
- Department of Medicine, University of New South Wales, Sydney, New South Wales, Australia
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
6
|
Zhang P, Huang P, Du J, He Y, Liu J, He G, Cui S, Zhang W, Li G, Chen S. Specific gut microbiota alterations in essential tremor and its difference from Parkinson's disease. NPJ Parkinsons Dis 2022; 8:98. [PMID: 35931717 PMCID: PMC9355955 DOI: 10.1038/s41531-022-00359-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/11/2022] [Indexed: 02/06/2023] Open
Abstract
Essential tremor (ET) is the most common movement disorder and share overlapping symptoms with Parkinson’s disease (PD), making differential diagnosis challenging. Gut dysbiosis is regarded crucial in the pathogenesis of PD. Since ET patients also has comorbidity in gastrointestinal disorders, the relationship between gut microbiota and ET really worth investigating and may help distinguishing ET from PD. Fecal samples from 54 ET, 67 de novo PD and 54 normal controls (NC) were collected for 16S ribosomal RNA gene sequencing and quantitative real-time PCR. ET showed lower species richness (Chao1 index) than NC and PD. ET was with Bacteroides-dominant enterotype, while PD was with Ruminococcus-dominant enterotype. Compared with NC, 7 genera were significantly reduced in ET, 4 of which (Ruminococcus, Romboutsia, Mucispirillum, and Aeromonas) were identified to be distinctive with an area under the curve (AUC) of 0.705. Compared to PD, 26 genera were found significantly different from ET, 4 of which (Bacteroides, Fusobacterium, Phascolarctobacterium, and Lachnospira) were found distinguishable with an AUC of 0.756. Clinical association results indicated that Proteus was associated with disease severity (TETRAS) of ET, while Klebsiella was linked to depression and anxiety in ET. Functional predictions revealed that 4 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were altered in ET. This study reveals gut dysbiosis in ET and it provides new insight into the pathogenesis of ET and helps distinguishing ET from PD.
Collapse
Affiliation(s)
- Pingchen Zhang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Pei Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Juanjuan Du
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Yixi He
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Jin Liu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Guiying He
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Shishuang Cui
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Weishan Zhang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Gen Li
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, People's Republic of China. .,Lab for Translational Research of Neurodegenerative Diseases, Shanghai Institute for Advanced Immunochemical Studies (SIAIS), Shanghai Tech University, Shanghai, 201210, People's Republic of China.
| |
Collapse
|
7
|
Huang W, Yau Y, Zhu J, Wang Y, Dai Z, Gan H, Qian L, Yang Z. Effect of Electroacupuncture at Zusanli (ST36) on Intestinal Microbiota in Rats With Chronic Atrophic Gastritis. Front Genet 2022; 13:824739. [PMID: 35281809 PMCID: PMC8906781 DOI: 10.3389/fgene.2022.824739] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/31/2022] [Indexed: 11/15/2022] Open
Abstract
Background: Electroacupuncture is a common treatment for chronic atrophic gastritis (CAG) in China. We aimed to determine the effects of electroacupuncture at zusanli (ST36) on intestinal microbiota in CAG rats. Methods: In total, 42 SD rats were randomly divided into normal (NC, 10 rats) and model (MG, 32 rats) groups. Rats in the MG group were established as CAG disease models. After that, the rats in the MG group were randomly divided into CAG (10 rats), electroacupuncture (EA, 10 rats), and Vitacoenzyme (Vit, 10 rats) groups. Rats in the NC and CAG groups were subjected to a 30-min/d confinement for 4 weeks. Rats in the EA group were given electroacupuncture at zusanli for 30 min/d for 4 weeks. Rats in the Vit group were given Vitacoenzyme solution 10 ml/(kg d) for 4 weeks. Histopathological changes in the gastric mucosa were observed with hematoxylin and eosin staining, and the gene expression level of p53, Bcl-2, and c-myc was determined using the qPCR method. The 16S rDNA sequencing technique was used to determine structural changes and relative abundance expression of intestinal flora. Results: Compared with the NC group, gastric mucosal pathology in the CAG group revealed significant inflammatory infiltration, and the gastric mucosal lesions in the electroacupuncture group were improved remarkably; the expression of p53 and c-myc genes in the CAG group increased (p < 0.05), while the expression of Bcl-2 genes decreased (p < 0.05) in the EA group, that of p53 and c-myc genes decreased (p < 0.05), and that of Bcl-2 genes increased (p < 0.05). The abundance of bacteria such as Lactobacillus, Desulfobacterota, and Bacteroides pectinophilus group in the CAG group increased (p < 0.05), while that of bacteria such as Gastranaerophilales, Romboutsia, and Blautia decreased (p < 0.05). The relative abundance of Desulfobacterota and Helicobacter in the EA group decreased (p < 0.05), while that of probiotic bacteria such as Oscillospirales, Romboutsia, and Christensenellaceae increased (p < 0.05). Conclusion: Electroacupuncture at zusanli can promote the repair of pathological damage to the gastric mucosa in rats with CAG, and the mechanism might relate to the reduction in the relative abundance of harmful bacteria, increase in the relative abundance of intestinal probiotics, and regulation of the intestinal microbiota.
Collapse
Affiliation(s)
- Wanyi Huang
- School of Medicine, Xiamen University, Xiamen, China.,College of Acupuncture and Moxibustion, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yuenming Yau
- School of Medicine, Xiamen University, Xiamen, China
| | - Jingru Zhu
- College of Acupuncture and Moxibustion, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yingjie Wang
- College of Acupuncture and Moxibustion, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zhipeng Dai
- Physical Education College, Hunan City University, Yiyang, China
| | - Huijuan Gan
- College of Acupuncture and Moxibustion, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Linchao Qian
- School of Medicine, Xiamen University, Xiamen, China
| | - Zongbao Yang
- School of Medicine, Xiamen University, Xiamen, China.,College of Acupuncture and Moxibustion, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
8
|
Potentially Probiotic Limosilactobacillus fermentum Fruit-Derived Strains Alleviate Cardiometabolic Disorders and Gut Microbiota Impairment in Male Rats Fed a High-Fat Diet. Probiotics Antimicrob Proteins 2022; 14:349-359. [PMID: 35066820 DOI: 10.1007/s12602-021-09889-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2021] [Indexed: 12/11/2022]
Abstract
High-fat diet (HFD) consumption is a risk factor for dyslipidemias, insulin resistance, and arterial hypertension linked with gut dysbiosis. Probiotic administration has been suggested as a safe therapeutic strategy for gut microbiota modulation and treatment and/or prevention of cardiometabolic disorders. Here, we assessed the effects of a potentially probiotic formulation containing strains of the Limosilactobacillus (L.) fermentum 139, 263, and 296 on the cardiometabolic disorders and gut microbiota derangements provoked by the HFD consumption. Male Wistar rats were allocated into control diet (CTL, n = 6), HFD (n = 6), and HFD receiving L. fermentum formulation (HFD-LF, n = 6) groups for 4 weeks. L. fermentum formulation (109 colony-forming unit (CFU)/ml of each strain) was daily administered by oral gavage. After 4-week follow-up, biochemical measurements, blood pressure (BP), heart rate (HR), sympathetic tone, and gut microbiota composition were evaluated. HFD consumption for 4 weeks increased lipid profile, insulin resistance, sympathetic tone, and blood pressure and impaired gut microbiota composition in male rats. Administration of L. fermentum formulation improved the gut microbiota composition, lipid profile, insulin resistance, autonomic dysfunction, and BP in rats fed with a HFD. Administration of a potentially fruit-derived probiotic formulation of L. fermentum strains improved gut microbiota composition and alleviated hyperlipidemia, insulin resistance, and sympathetic hyperactivity and increased BP in rats fed a HFD. Our findings may encourage the development of randomized controlled trials to assess the effects of L. fermentum treatment in subjects with cardiometabolic disorders.
Collapse
|
9
|
Salillas S, Galano-Frutos JJ, Mahía A, Maity R, Conde-Giménez M, Anoz-Carbonell E, Berlamont H, Velazquez-Campoy A, Touati E, Mamat U, Schaible UE, Gálvez JA, Díaz-de-Villegas MD, Haesebrouck F, Aínsa JA, Sancho J. Selective Targeting of Human and Animal Pathogens of the Helicobacter Genus by Flavodoxin Inhibitors: Efficacy, Synergy, Resistance and Mechanistic Studies. Int J Mol Sci 2021; 22:ijms221810137. [PMID: 34576300 PMCID: PMC8467567 DOI: 10.3390/ijms221810137] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/13/2021] [Accepted: 09/16/2021] [Indexed: 02/08/2023] Open
Abstract
Antimicrobial resistant (AMR) bacteria constitute a global health concern. Helicobacter pylori is a Gram-negative bacterium that infects about half of the human population and is a major cause of peptic ulcer disease and gastric cancer. Increasing resistance to triple and quadruple H. pylori eradication therapies poses great challenges and urges the development of novel, ideally narrow spectrum, antimicrobials targeting H. pylori. Here, we describe the antimicrobial spectrum of a family of nitrobenzoxadiazol-based antimicrobials initially discovered as inhibitors of flavodoxin: an essential H. pylori protein. Two groups of inhibitors are described. One group is formed by narrow-spectrum compounds, highly specific for H. pylori, but ineffective against enterohepatic Helicobacter species and other Gram-negative or Gram-positive bacteria. The second group includes extended-spectrum antimicrobials additionally targeting Gram-positive bacteria, the Gram-negative Campylobacter jejuni, and most Helicobacter species, but not affecting other Gram-negative pathogens. To identify the binding site of the inhibitors in the flavodoxin structure, several H. pylori-flavodoxin variants have been engineered and tested using isothermal titration calorimetry. An initial study of the inhibitors capacity to generate resistances and of their synergism with antimicrobials commonly used in H. pylori eradication therapies is described. The narrow-spectrum inhibitors, which are expected to affect the microbiota less dramatically than current antimicrobial drugs, offer an opportunity to develop new and specific H. pylori eradication combinations to deal with AMR in H. pylori. On the other hand, the extended-spectrum inhibitors constitute a new family of promising antimicrobials, with a potential use against AMR Gram-positive bacterial pathogens.
Collapse
Affiliation(s)
- Sandra Salillas
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - Juan José Galano-Frutos
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - Alejandro Mahía
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - Ritwik Maity
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - María Conde-Giménez
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
| | - Ernesto Anoz-Carbonell
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
| | - Helena Berlamont
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B9820 Merelbeke, Belgium; (H.B.); (F.H.)
| | - Adrian Velazquez-Campoy
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- ARAID Foundation, Government of Aragon, 50018 Zaragoza, Spain
- CIBER de Enfermedades Hepáticas y Digestivas CIBERehd, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Eliette Touati
- Unit of Helicobacter Pathogenesis, CNRS UMR2001, Department of Microbiology, Institut Pasteur, 25-28 Rue du Dr. Roux, 75724 Paris, France;
| | - Uwe Mamat
- Cellular Microbiology, Program Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany; (U.M.); (U.E.S.)
| | - Ulrich E. Schaible
- Cellular Microbiology, Program Area Infections, Research Center Borstel, Leibniz Lung Center, 23845 Borstel, Germany; (U.M.); (U.E.S.)
| | - José A. Gálvez
- Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC—Departamento de Química Orgánica, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain; (J.A.G.); (M.D.D.-d.-V.)
| | - María D. Díaz-de-Villegas
- Instituto de Síntesis Química y Catálisis Homogénea (ISQCH), CSIC—Departamento de Química Orgánica, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain; (J.A.G.); (M.D.D.-d.-V.)
| | - Freddy Haesebrouck
- Department of Pathobiology, Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, B9820 Merelbeke, Belgium; (H.B.); (F.H.)
| | - José A. Aínsa
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Faculty of Medicine, University of Zaragoza, 50009 Zaragoza, Spain
- CIBER de Enfermedades Respiratorias—CIBERES, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Sancho
- Biocomputation and Complex Systems Physics Institute (BIFI)-Joint Units: BIFI-IQFR (CSIC) and GBsC-CSIC, University of Zaragoza, 50018 Zaragoza, Spain; (S.S.); (J.J.G.-F.); (A.M.); (R.M.); (M.C.-G.); (E.A.-C.); (A.V.-C.); (J.A.A.)
- Departamento de Bioquímica y Biología Molecular y Celular, Faculty of Science, University of Zaragoza, 50009 Zaragoza, Spain
- Aragon Health Research Institute (IIS Aragón), 50009 Zaragoza, Spain
- Correspondence:
| |
Collapse
|
10
|
Ochoa S, Collado L. Enterohepatic Helicobacter species - clinical importance, host range, and zoonotic potential. Crit Rev Microbiol 2021; 47:728-761. [PMID: 34153195 DOI: 10.1080/1040841x.2021.1924117] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The genus Helicobacter defined just over 30 years ago, is a highly diverse and fast-growing group of bacteria that are able to persistently colonize a wide range of animals. The members of this genus are subdivided into two groups with different ecological niches, associated pathologies, and phylogenetic relationships: the gastric Helicobacter (GH) and the enterohepatic Helicobacter (EHH) species. Although GH have been mostly studied, EHH species have become increasingly important as emerging human pathogens and potential zoonotic agents in the last years. This group of bacteria has been associated with the development of several diseases in humans from acute pathologies like gastroenteritis to chronic pathologies that include inflammatory bowel disease, and liver and gallbladder diseases. However, their reservoirs, as well as their routes of transmission, have not been well established yet. Therefore, this review summarizes the current knowledge of taxonomy, epidemiology, and clinical role of the EHH group.
Collapse
Affiliation(s)
- Sofia Ochoa
- Faculty of Sciences, Institute of Biochemistry and Microbiology, Universidad Austral de Chile, Valdivia, Chile.,ANID - Millennium Science Initiative Program - Millennium Nucleus in the Biology of the Intestinal Microbiota, Santiago, Chile
| | - Luis Collado
- Faculty of Sciences, Institute of Biochemistry and Microbiology, Universidad Austral de Chile, Valdivia, Chile.,ANID - Millennium Science Initiative Program - Millennium Nucleus in the Biology of the Intestinal Microbiota, Santiago, Chile
| |
Collapse
|
11
|
Axarlis K, Daskalaki MG, Michailidou S, Androulaki N, Tsoureki A, Mouchtaropoulou E, Kolliniati O, Lapi I, Dermitzaki E, Venihaki M, Kousoulaki K, Argiriou A, Marsni ZE, Tsatsanis C. Diet Supplementation with Fish-Derived Extracts Suppresses Diabetes and Modulates Intestinal Microbiome in a Murine Model of Diet-Induced Obesity. Mar Drugs 2021; 19:268. [PMID: 34064922 PMCID: PMC8151984 DOI: 10.3390/md19050268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 04/29/2021] [Accepted: 05/07/2021] [Indexed: 02/07/2023] Open
Abstract
Metabolic syndrome-related diseases affect millions of people worldwide. It is well established that changes in nutritional habits and lifestyle can improve or prevent metabolic-related pathologies such as type-2 diabetes and obesity. Previous reports have shown that nutritional supplements have the capacity to limit glucose intolerance and suppress diabetes development. In this study, we investigated the effect of dietary supplementation with fish-derived extracts on obesity and type 2 diabetes and their impact on gut microbial composition. We showed that nutritional supplements containing Fish Complex (FC), Fish Complex combined with Cod Powder (FC + CP), or Cod Powder combined with Collagen (CP + C) improved glucose intolerance, independent of abdominal fat accumulation, in a mouse model of diet-induced obesity and type 2 diabetes. In addition, collagen-containing supplements distinctly modulate the gut microbiome in high-fat induced obesity in mice. Our results suggest that fish-derived supplements suppress diet-induced type 2 diabetes, which may be partly mediated through changes in the gut microbiome. Thus, fish-derived supplements and particularly the ones containing fish collagen have potential beneficial properties as dietary supplements in managing type 2 diabetes and metabolic syndrome via modulation of the gut microbiome.
Collapse
Affiliation(s)
- Konstantinos Axarlis
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (K.A.); (M.G.D.); (N.A.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Maria G. Daskalaki
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (K.A.); (M.G.D.); (N.A.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Sofia Michailidou
- Institute of Applied Biosciences (INAB), CERTH, Thermi, 57001 Thessaloniki, Greece; (S.M.); (A.T.); (E.M.); (A.A.)
| | - Nikolais Androulaki
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (K.A.); (M.G.D.); (N.A.); (O.K.); (I.L.); (E.D.); (M.V.)
| | - Antiopi Tsoureki
- Institute of Applied Biosciences (INAB), CERTH, Thermi, 57001 Thessaloniki, Greece; (S.M.); (A.T.); (E.M.); (A.A.)
| | - Evangelia Mouchtaropoulou
- Institute of Applied Biosciences (INAB), CERTH, Thermi, 57001 Thessaloniki, Greece; (S.M.); (A.T.); (E.M.); (A.A.)
| | - Ourania Kolliniati
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (K.A.); (M.G.D.); (N.A.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Ioanna Lapi
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (K.A.); (M.G.D.); (N.A.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Eirini Dermitzaki
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (K.A.); (M.G.D.); (N.A.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| | - Maria Venihaki
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (K.A.); (M.G.D.); (N.A.); (O.K.); (I.L.); (E.D.); (M.V.)
| | - Katerina Kousoulaki
- Department of Nutrition and Feed Technology, Nofima AS, 5141 Bergen, Norway;
| | - Anagnostis Argiriou
- Institute of Applied Biosciences (INAB), CERTH, Thermi, 57001 Thessaloniki, Greece; (S.M.); (A.T.); (E.M.); (A.A.)
- Department of Food Science and Nutrition, University of the Aegean, Myrina, 81400 Lemnos, Greece
| | | | - Christos Tsatsanis
- Laboratory of Clinical Chemistry, Medical School, University of Crete, 70013 Heraklion, Greece; (K.A.); (M.G.D.); (N.A.); (O.K.); (I.L.); (E.D.); (M.V.)
- Institute of Molecular Biology and Biotechnology, FORTH, 71100 Heraklion, Greece
| |
Collapse
|
12
|
Matos R, Amorim I, Magalhães A, Haesebrouck F, Gärtner F, Reis CA. Adhesion of Helicobacter Species to the Human Gastric Mucosa: A Deep Look Into Glycans Role. Front Mol Biosci 2021; 8:656439. [PMID: 34026832 PMCID: PMC8138122 DOI: 10.3389/fmolb.2021.656439] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/22/2021] [Indexed: 12/15/2022] Open
Abstract
Helicobacter species infections may be associated with the development of gastric disorders, such as gastritis, peptic ulcers, intestinal metaplasia, dysplasia and gastric carcinoma. Binding of these bacteria to the gastric mucosa occurs through the recognition of specific glycan receptors expressed by the host epithelial cells. This review addresses the state of the art knowledge on these host glycan structures and the bacterial adhesins involved in Helicobacter spp. adhesion to gastric mucosa colonization. Glycans are expressed on every cell surface and they are crucial for several biological processes, including protein folding, cell signaling and recognition, and host-pathogen interactions. Helicobacter pylori is the most predominant gastric Helicobacter species in humans. The adhesion of this bacterium to glycan epitopes present on the gastric epithelial surface is a crucial step for a successful colonization. Major adhesins essential for colonization and infection are the blood-group antigen-binding adhesin (BabA) which mediates the interaction with fucosylated H-type 1 and Lewis B glycans, and the sialic acid-binding adhesin (SabA) which recognizes the sialyl-Lewis A and X glycan antigens. Since not every H. pylori strain expresses functional BabA or SabA adhesins, other bacterial proteins are most probably also involved in this adhesion process, including LabA (LacdiNAc-binding adhesin), which binds to the LacdiNAc motif on MUC5AC mucin. Besides H. pylori, several other gastric non-Helicobacter pylori Helicobacters (NHPH), mainly associated with pigs (H. suis) and pets (H. felis, H. bizzozeronii, H. salomonis, and H. heilmannii), may also colonize the human stomach and cause gastric disease, including gastritis, peptic ulcers and mucosa-associated lymphoid tissue (MALT) lymphoma. These NHPH lack homologous to the major known adhesins involved in colonization of the human stomach. In humans, NHPH infection rate is much lower than in the natural hosts. Differences in the glycosylation profile between gastric human and animal mucins acting as glycan receptors for NHPH-associated adhesins, may be involved. The identification and characterization of the key molecules involved in the adhesion of gastric Helicobacter species to the gastric mucosa is important to understand the colonization and infection strategies displayed by different members of this genus.
Collapse
Affiliation(s)
- Rita Matos
- Instituto de Investigação e Inovação Em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto (ICBAS), Porto, Portugal
| | - Irina Amorim
- Instituto de Investigação e Inovação Em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto (ICBAS), Porto, Portugal
| | - Ana Magalhães
- Instituto de Investigação e Inovação Em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal
| | - Freddy Haesebrouck
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Ghent, Belgium
| | - Fátima Gärtner
- Instituto de Investigação e Inovação Em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal
| | - Celso A. Reis
- Instituto de Investigação e Inovação Em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto (ICBAS), Porto, Portugal
- Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| |
Collapse
|
13
|
Rasmussen SL, Ørsted I, Tarpgaard IH, Nielsen HL. Helicobacter cinaedi bacteraemia secondary to enterocolitis in an immunocompetent patient. Gut Pathog 2021; 13:26. [PMID: 33888153 PMCID: PMC8063464 DOI: 10.1186/s13099-021-00422-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/12/2021] [Indexed: 01/13/2023] Open
Abstract
Background Helicobacter cinaedi are motile, gram-negative spiral rods with a natural reservoir in the intestinal tract of hamsters and rhesus monkeys. In humans, H. cinaedi has been reported in different human infections like fever, abdominal pain, gastroenteritis, proctitis, diarrhoea, erysipelas, cellulitis, arthritis, and neonatal meningitis typically diagnosed by positive blood cultures. Even though H. cinaedi has been detected from human blood and stool the entry of H. cinaedi into the blood stream was undocumented until quite recently. The use of pulse-field gel electrophoresis (PFGE) demonstrated that stool- and blood-derived H. cinaedi strains were consistent. Case presentation Here, we describe a rare Danish case of H. cinaedi bacteraemia in an immunocompetent 44-year-old male with diarrhoea. We isolated H. cinaedi from a blood culture taken at admission, and from a FecalSwab taken at day six despite ongoing antibiotic therapy. Next, we made a genetic comparison of both isolates by use of Multi-locus sequence typing (MLST)- and Single nucleotide polymorphism (SNP)-analysis. The two isolates were identical with zero SNPs and by use of MLST the isolate was identified as a novel ST20, confirming previous data of the intestinal tract as a route of H. cinaedi bacteraemia. The results of our AST showed a resistance pattern with higher MICs for ciprofloxacin and clarithromycin than for ampicillin, amoxicillin, gentamicin, and imipenem. The patient was cured with targeted therapy with pivampicillin; however, the primary source of transmission was unknown. Conclusions In conclusion, this case of H. cinaedi bacteraemia secondary to enterocolitis in an immunocompetent patient provide clear evidence that one route of infection occurs through translocation from the intestinal tract to the bloodstream. Helicobacter cinaedi from blood and faeces were identical with a novel ST20, resistant to ciprofloxacin and clarithromycin however, the patient was cured with oral pivampicillin.
Collapse
Affiliation(s)
- Sofie Larsen Rasmussen
- Department of Clinical Microbiology, Aalborg University Hospital, Hobrovej 18, 9000, Aalborg, Denmark
| | - Iben Ørsted
- Department of Infectious Diseases, Aalborg University Hospital, Hobrovej 18, 9000, Aalborg, Denmark
| | - Irene Harder Tarpgaard
- Department of Clinical Microbiology, Aalborg University Hospital, Hobrovej 18, 9000, Aalborg, Denmark
| | - Hans Linde Nielsen
- Department of Clinical Microbiology, Aalborg University Hospital, Hobrovej 18, 9000, Aalborg, Denmark. .,Department of Clinical Medicine, Aalborg University, Aalborg, Denmark.
| |
Collapse
|
14
|
Helicobacter canis bacteraemia in a rheumatoid arthritis patient treated with tofacitinib: case report and literature review. Ann Clin Microbiol Antimicrob 2021; 20:22. [PMID: 33827581 PMCID: PMC8028780 DOI: 10.1186/s12941-021-00426-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Non-Helicobacter pylori species (NHPS) are newly emerging bacteria that naturally inhabit birds and mammals apart from humans and rarely cause diseases in humans. In recent years, a rise in the number of cases associated with NHPS infections in humans has been observed. Among them, infections with Helicobacter (H.) canis are sporadic and challenging to recognise clinically. To date, ten cases of H. canis infections in mainly immunocompromised humans have been reported in the literature. Transmission pathway is most likely zoonotic via the faecal-oral route during close contacts with dogs and cats or may result from a contaminated sheep milk intake. No clear guidelines for successful antibiotic regimen are known. Important additional risk factor for infection might be biologic agents and Janus kinase inhibitors (JAKi) used in the treatment of rheumatoid arthritis (RA) and other conditions. Herein we present the first case of H. canis bacteraemia in a RA patient treated with novel JAKi tofacitinib. CASE PRESENTATION A 65-year-old female patient with RA and rituximab-induced hypogammaglobulinemia treated with tofacitinib, methotrexate, and methylprednisolone came to a planned visit in our outpatient rheumatology clinic. She presented with a history of back pain that significantly worsened 2 days before visit. She had numbness and tingling sensation in both legs and muscle weakness. Neurological examination was within a normal range. The patient was afebrile, had no chills, and was haemodynamically stable. She was in close contact with her pet dogs. Laboratory examination showed increased markers of inflammation. She was found to have H. canis bacteraemia with underlying multilevel degenerative lumbar spinal stenosis. Identification of H. canis was performed by MALDI-TOF MS and 16 S rRNA gene sequence analysis of isolate from subcultured positive aerobic blood culture bottles. Antimicrobial susceptibility testing showed low minimum inhibitory concentrations to amoxicillin-clavulanate, cefotaxime, ceftriaxone, meropenem, and gentamicin. She was treated with combined antibiotic regimen (ceftriaxone, doxycycline) for 14 days, which resulted in total remission of the infection. CONCLUSIONS Clinicians should recognise H. canis infection risk in patients with recent pet exposure and predisposing factors such as immunodeficiency disorders or diseases that demand immunosuppressive drug therapy. A minimum of two weeks of antibiotic therapy is suggested.
Collapse
|
15
|
van den Brule S, Rappe M, Ambroise J, Bouzin C, Dessy C, Paquot A, Muccioli GG, Lison D. Diesel exhaust particles alter the profile and function of the gut microbiota upon subchronic oral administration in mice. Part Fibre Toxicol 2021; 18:7. [PMID: 33563307 PMCID: PMC7871568 DOI: 10.1186/s12989-021-00400-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/29/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Ambient air pollution by particulate matters, including diesel exhaust particles (DEP), is a major cause of cardiovascular and metabolic mortality worldwide. The mechanisms by which DEP cause these adverse outcomes are not completely understood. Because the gut microbiota controls cardiovascular and metabolic health, we hypothesized that the fraction of inhaled DEP which reach the gut after mucociliary clearance and swallowing might induce gut dysbiosis and, in turn, contribute to aggravate or induce cardiovascular and metabolic diseases. RESULTS Female ApoE-/- mice fed a Western diet, and wild-type (C57Bl/6) mice fed standard diet were gavaged with DEP (SRM2975) doses corresponding to mucociliary clearance from inhalation exposure (200 or 1000 ng/day, 3 times a week for 3 months; and 40, 200 or 1000 ng/day, 3 times a week for 6 months, respectively). No mortality, overt systemic or digestive toxicity was observed. A dose-dependent alteration of the gut microbiota was recorded in both strains. In ApoE-/-, β-diversity was modified by DEP, but no significant modification of the relative abundance of the phyla, families or genera was identified. In C57BL/6 mice, DEP reduced α-diversity (Shannon and Simpson indices), and modified β-diversity, including a reduction of the Proteobacteria and Patescibacteria phyla, and an increase of the Campylobacterota phylum. In both mouse models, perturbation of the gut microbiota composition was associated with a dose-dependent reduction of bacterial short chain fatty acids (butyrate and propionate) in cecal content. However, DEP ingestion did not aggravate (ApoE-/-), or induce (C57BL/6 mice) atherosclerotic plaques, and no metabolic alteration (glucose tolerance, resistance to insulin, or lipidemia) was recorded. CONCLUSIONS We show here that oral exposure to DEP, at doses relevant for human health, changes the composition and function of the gut microbiota. These modifications were, however, not translated into ultimate atherosclerotic or metabolic outcomes.
Collapse
Affiliation(s)
- Sybille van den Brule
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Margaux Rappe
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Jérôme Ambroise
- Centre de Technologies Moléculaires Appliquées, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Caroline Bouzin
- IREC Imaging Platform (2IP), Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
| | - Chantal Dessy
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique, UCLouvain and Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Adrien Paquot
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids Research Group, Louvain Drug Research Institute, UCLouvain, Brussels, Belgium
| | - Dominique Lison
- Louvain centre for Toxicology and Applied Pharmacology, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium.
| |
Collapse
|
16
|
Fox-Lewis A, Basu I, Vesty A, Henderson G, Chhibber AV, Thomas M. Helicobacter cinaedi bacteremia in a returning traveler. IDCases 2020; 21:e00910. [PMID: 32695610 PMCID: PMC7365975 DOI: 10.1016/j.idcr.2020.e00910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 11/29/2022] Open
Abstract
Of the non‐Helicobacter pylori Helicobacter (NHPH) species, Helicobacter cinaedi is an emerging cause of infection in humans. Here we report a novel clinical presentation of H. cinaedi infection: a case of fever in a returning traveler. A 31 year old previously fit and well male presented with onset of fever 24 h after returning from travel in Singapore and Indonesia. Associated symptoms consisted of sore throat, mild shortness of breath, generalized myalgia and arthralgia, headache, and four episodes of loose stools. The patient recovered spontaneously without treatment and was discharged. After 4 days of incubation, blood cultures grew H. cinaedi. H. cinaedi is a slow-growing fastidious organism poorly detected by some commonly used automated blood culture systems, and difficult to identify using commercial or traditional biochemical identification systems. This case illustrates the importance of H. cinaedi as an emerging pathogen in immunocompetent patients, with a wide variety of possible clinical presentations. The challenges in the microbiological diagnosis of H. cinaedi infections lead us to speculate that H. cinaedi is an underdiagnosed cause of febrile illness, both in returning travelers and in other clinical settings.
Collapse
Affiliation(s)
- Andrew Fox-Lewis
- Microbiology Department, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - Indira Basu
- Microbiology Department, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - Anna Vesty
- Microbiology Department, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - Gillian Henderson
- Microbiology Department, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - Aakash V Chhibber
- Microbiology Department, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - Mark Thomas
- Infectious Diseases Department, Auckland City Hospital, Auckland, New Zealand
| |
Collapse
|