1
|
Pongsupasamit P, Thonusin C, Luewan S, Chattipakorn N, Chattipakorn SC. Beyond hormones: 3PM approach to vaginal microbiota dynamics in postmenopausal women. EPMA J 2025; 16:299-350. [PMID: 40438491 PMCID: PMC12106263 DOI: 10.1007/s13167-025-00406-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 03/12/2025] [Indexed: 06/01/2025]
Abstract
Menopause marks a critical transition characterized by ceased ovarian function and declining estrogen levels, affecting multiple systems with vasomotor symptoms and genitourinary syndrome of menopause (GSM). Recent evidence shows vaginal microbiota undergoes significant alterations during menopause, influencing GSM severity. This comprehensive review examined vaginal microbiota dynamics in postmenopausal women through Predictive, Preventive, and Personalized Medicine (3PM/PPPM), revealing characteristic shifts-increased alpha diversity, reduced Lactobacillus dominance, and transitions toward non-Lactobacillus species-that serve as potential predictive biomarkers for the menopausal state, premature ovarian insufficiency, and GSM symptoms. The analysis evaluated microbiota-based risk stratification strategies for vaginal dysbiosis and demonstrated the effectiveness of both hormonal interventions (systemic/local estrogen, tibolone, ospemifene) and non-hormonal alternatives (probiotics, energy-based devices, pessary) in normalizing microbiota composition and improving vaginal health. The application of PPPM/3PM transformed menopausal healthcare from reactive to proactive precision-based care by establishing microbiota-based biomarkers that predict health risks, enable early targeted interventions tailored to specific microbiota profiles, and guide personalized treatment approaches based on individual microbial compositions. While this paradigm shift significantly advances gynecological medicine, research gaps remain in validating baseline microbiota signatures as predictive biomarkers and establishing standardized screening protocols. Further studies are needed to validate interventions such as probiotics and prebiotics, optimizing strain selection for personalized, evidence-based preventive and therapeutic strategies. Developing standardized yet personalized protocols to restore a balanced vaginal microbiome could help alleviate menopause-related symptoms. Advancing microbiota-based personalized therapeutic approaches is crucial to enhancing the quality of life for postmenopausal women through targeted and individualized vaginal health management.
Collapse
Affiliation(s)
- Panchita Pongsupasamit
- Division of Female Pelvic Medicine and Reconstructive Surgery, Department of Obstetrics and Gynecology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chanisa Thonusin
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Suchaya Luewan
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- The Academy of Science, the Royal Society of Thailand, Bangkok, Thailand
| | - Siriporn C. Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200 Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200 Thailand
| |
Collapse
|
2
|
Du X, Liu M, Li J, Liu Y, Ge S, Gao H, Zhang M. Bifidobacterium animalis Supplementation Improves Intestinal Barrier Function and Alleviates Antibiotic-Associated Diarrhea in Mice. Foods 2025; 14:1704. [PMID: 40428484 PMCID: PMC12110814 DOI: 10.3390/foods14101704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2025] [Revised: 05/09/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Probiotics have gained increasing recognition for their potential to mitigate antibiotic-associated diarrhea (AAD). However, the precise mechanisms underlying their effects remain unclear. This study developed a mouse model of AAD using ceftriaxone to investigate the alleviating effects and mechanisms of Bifidobacterium animalis A6 (A6). The findings indicated that A6 supplementation effectively attenuated ceftriaxone-associated diarrhea in mice. The morphological damage to the villi and crypts was partially restored and more neatly reorganized following the A6 intervention. Additionally, intestinal morphology observations revealed a significant increase in the thickness of the mucus layer in the A6-treated group. Further examination of key regulatory genes associated with mucus secretion demonstrated that the A6 intervention effectively upregulated the expression of mucin1, thereby reinforcing the mucus layer. Concurrently, the A6 intervention upregulated the expression of the AQP4 and SLC26A3 genes in the intestine, which is responsible for restoring water absorption capacity in AAD mice. Additionally, the A6 treatment reduced ceftriaxone-induced harm to the intestinal microbiota of the mice, boosting beneficial bacteria like Bacteroidales, Akkermansia, Bifidobacterium, and Lactobacillus. Overall, this study offers valuable insights into the potential therapeutic role of A6 in restoring intestinal homeostasis and alleviating symptoms associated with AAD.
Collapse
Affiliation(s)
- Xiaoyu Du
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Mingkun Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230000, China
| | - Jingyu Li
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Yue Liu
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Shaoyang Ge
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Haina Gao
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Ming Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China
| |
Collapse
|
3
|
Holcomb M, Marshall AG, Flinn H, Lozano-Cavazos M, Soriano S, Gomez-Pinilla F, Treangen TJ, Villapol S. Probiotic treatment induces sex-dependent neuroprotection and gut microbiome shifts after traumatic brain injury. J Neuroinflammation 2025; 22:114. [PMID: 40254574 PMCID: PMC12010691 DOI: 10.1186/s12974-025-03419-1] [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: 03/31/2024] [Accepted: 03/16/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Recent studies have highlighted the potential influence of gut dysbiosis on traumatic brain injury (TBI) outcomes. Alterations in the abundance and diversity of Lactobacillus species may affect immune dysregulation, neuroinflammatory responses, anxiety- and depressive-like behaviors, and neuroprotective mechanisms activated in response to TBI. OBJECTIVE This study aims to evaluate the protective and preventive effects of Pan-probiotic (PP) treatment on the inflammatory response during both the acute and chronic phases of TBI. METHODS Males and female mice underwent controlled cortical impact (CCI) injury or sham. They received a PP mixture in drinking water containing strains of Lactobacillus plantarum, L. reuteri, L. helveticas, L. fermentum, L. rhamnosus, L. gasseri, and L. casei. In the acute group, mice received PP or vehicle (VH) treatment for 7 weeks before TBI, continuing until 3 days post-injury (dpi). In the chronic group, treatment began 2 weeks before TBI and was extended through 35 dpi. The taxonomic microbiome profiles of fecal samples were evaluated using 16S rRNA V1-V3 sequencing analysis, and Short-chain fatty acids (SCFAs) were measured. Immunohistochemical, in situ hybridization, and histological analyses were performed to assess neuroinflammation post-TBI, while behavioral assessments were conducted to evaluate sensorimotor and cognitive functions. RESULTS Our findings suggest that a 7-week PP administration induces specific microbial changes, including increased abundance of beneficial bacteria such as Lactobacillaceae, Limosilactobacillus, and Lactiplantibacillus. PP treatment reduces lesion volume and cell death at 3 dpi, elevates SCFA levels at 35 dpi, and decreases microglial activation at both time points, particularly in males. Additionally, PP treatment improved motor recovery in males and alleviated depressive-like behaviors in females. CONCLUSION Our findings indicate that PP administration modulates microbiome composition, reduces neuroinflammation, and improves motor deficits following TBI, with these effects being particularly pronounced in male mice.
Collapse
Affiliation(s)
- Morgan Holcomb
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Austin G Marshall
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
- Department of Computer Science, Rice University, Houston, TX, USA
| | - Hannah Flinn
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Mariana Lozano-Cavazos
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Sirena Soriano
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA
| | - Fernando Gomez-Pinilla
- Departments of Neurosurgery and Integrative Biology and Physiology, Brain Injury Research Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Todd J Treangen
- Department of Computer Science, Rice University, Houston, TX, USA
- Department of Bioengineering, Rice University, Houston, TX, USA
- Ken Kennedy Institute, Rice University, Houston, TX, USA
| | - Sonia Villapol
- Department of Neurosurgery and Center for Neuroregeneration, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Bioengineering, Rice University, Houston, TX, USA.
- Department of Neuroscience in Neurological Surgery, Weill Cornell Medical College, New York City, NY, USA.
| |
Collapse
|
4
|
Gomez-Pinilla F, Myers SK. Traumatic brain injury from a peripheral axis perspective: Uncovering the roles of liver and adipose tissue in temperature regulation. Prog Neurobiol 2025; 247:102733. [PMID: 40032155 DOI: 10.1016/j.pneurobio.2025.102733] [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: 11/06/2024] [Revised: 01/21/2025] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
Traumatic brain injury (TBI) is a major cause of death and disability worldwide. Most current treatments for TBI and other neurological disorders focus on the brain, often overlooking the significant contributions of peripheral organs to disease progression. Emerging evidence suggests that organs such as the liver and adipose tissue play crucial roles in TBI pathogenesis. The liver synthesizes lipids and proteins vital for brain function, while adipose tissue provides hormones and metabolites that influence brain activity. New research indicates that the liver and adipose tissue work in concert with the hypothalamus to regulate essential processes, such as body temperature, which become disrupted in TBI. Additionally, the brain-peripheral axis-a complex network of visceral nerve pathways, hormones, and metabolites-plays a bidirectional role in regulating brain plasticity and function. Understanding how TBI leads to dysregulation of the liver, adipose tissue, and other organs could unlock new therapeutic opportunities for treating TBI and related neurological disorders. The intricate autonomic network involving hypothalamic and enteric neurons, along with visceral nerve pathways and hormones, presents both pathological targets and therapeutic potential. We examine scientific evidence suggesting that correcting disturbances in systemic physiology could enhance the brain's capacity for healing. However, the interdependence of this autonomic network implies that treating dysfunction in one area may affect others. Therefore, we also explore the mechanisms by which diet and exercise can comprehensively impact the brain-peripheral axis, supporting the healing process. CHEMICAL COMPOUNDS: D-Fructose (PubChem CID 2723872); docosahexaenoic acid (PubChem CID 45934466); eicosapentaenoic acid (PubChem 5282847).
Collapse
Affiliation(s)
- F Gomez-Pinilla
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA 90095, USA; Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA.
| | - Sydney K Myers
- Department of Neurosurgery, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Hou X, Wang M, Hu T, Wu Z, Liang H, Zhong Y, Ma Z, Zhang H, Xiao L, Zhang W, Zou Y. Evaluation of the safety and probiotic properties of Limosilactobacillus fermentum BGI-AF16, a uric acid-lowering probiotic strain. Microb Pathog 2025; 201:107382. [PMID: 39961375 DOI: 10.1016/j.micpath.2025.107382] [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: 05/06/2024] [Revised: 01/08/2025] [Accepted: 02/15/2025] [Indexed: 02/23/2025]
Abstract
Some beneficial microorganisms in the intestine have the potential to degrade uric acid, offering a novel strategy for the prevention of hyperuricemia. In this study, the safety and probiotic potentials of Limosilactobacillus fermentum BGI-AF16 were evaluated by whole genome sequence analysis and in vitro experiments. Based on the gene analysis of antibiotic resistance and virulence factors, L. fermentum BGI-AF16 has been shown to be safe. We identified probiotic-related genes by genome annotation tools and conducted in vitro experiments to evaluate the ability of L. fermentum BGI-AF16 to inhibit pathogenic bacteria, tolerate a simulated gastrointestinal environment, and degrade uric acid. The results from in vitro experiments showed that L. fermentum BGI-AF16 had inhibitory effects on four clinically relevant pathogens and was highly tolerant to the gastrointestinal environment. In addition, L. fermentum BGI-AF16 was able to rapidly degrade uric acid within the first hour, and the strain could degrade 56.36 ± 2.32 % of uric acid by the third hour. The genome of the strain contains genes encoding flavin adenine dinucleotide (FAD)-dependent urate hydroxylase (EC.1.14.13.113), an enzyme that directly metabolizes uric acid. And the strain has a complete uric acid metabolic pathway. These results suggest that L. fermentum BGI-AF16 is a probiotic candidate with significant potential for reducing uric acid level.
Collapse
Affiliation(s)
- Xiaoxue Hou
- BGI Research, Shenzhen, 518083, China; College of Agriculture, South China Agricultural University, Guangzhou, 510642, China
| | - Mengmeng Wang
- BGI Research, Shenzhen, 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | - Zhinan Wu
- BGI Research, Shenzhen, 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | | | - Yiyi Zhong
- BGI Precision Nutrition, Shenzhen, 518083, China
| | - Zhihui Ma
- BGI Precision Nutrition, Shenzhen, 518083, China
| | | | - Liang Xiao
- BGI Research, Shenzhen, 518083, China; Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, 518083, China
| | - Wenjin Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China; BGI Genomics, Shenzhen, 518083, China
| | - Yuanqiang Zou
- BGI Research, Shenzhen, 518083, China; Shenzhen Engineering Laboratory of Detection and Intervention of Human Intestinal Microbiome, BGI Research, Shenzhen, 518083, China.
| |
Collapse
|
6
|
Chen A, Gong Y, Wu S, Du Y, Liu Z, Jiang Y, Li J, Miao YB. Navigating a challenging path: precision disease treatment with tailored oral nano-armor-probiotics. J Nanobiotechnology 2025; 23:72. [PMID: 39893419 PMCID: PMC11786591 DOI: 10.1186/s12951-025-03141-3] [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: 10/23/2024] [Accepted: 01/19/2025] [Indexed: 02/04/2025] Open
Abstract
Oral probiotics have significant potential for preventing and treating many diseases. Yet, their efficacy is often hindered by challenges related to survival and colonization within the gastrointestinal tract. Nanoparticles emerge as a transformative solution, offering robust protection and enhancing the stability and bioavailability of these probiotics. This review explores the innovative application of nanoparticle-armored engineered probiotics for precise disease treatment, specifically addressing the physiological barriers associated with oral administration. A comprehensive evaluation of various nano-armor probiotics and encapsulation methods is provided, carefully analyzing their respective merits and limitations, alongside strategies to enhance probiotic survival and achieve targeted delivery and colonization within the gastrointestinal tract. Furthermore, the review explores the potential clinical applications of nano-armored probiotics in precision therapeutics, critically addressing safety and regulatory considerations, and proposing the innovative concept of 'probiotic intestinal colonization with nano armor' for brain-targeted therapies. Ultimately, this review aspires to guide the advancement of nano-armored probiotic therapies, driving progress in precision medicine and paving the way for groundbreaking treatment modalities.
Collapse
Affiliation(s)
- Anmei Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Ying Gong
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Shaoquan Wu
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Ye Du
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China
| | - Zhijun Liu
- Urology Institute of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, 518000, China
| | - Yuhong Jiang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China.
| | - Jiahong Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610041, China.
| | - Yang-Bao Miao
- Department of Haematology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu, 610000, China.
| |
Collapse
|
7
|
Nami Y, Barghi A, Shahgolzari M, Salehian M, Haghshenas B. Mechanism of Action and Beneficial Effects of Probiotics in Amateur and Professional Athletes. Food Sci Nutr 2025; 13:e4658. [PMID: 39803224 PMCID: PMC11717059 DOI: 10.1002/fsn3.4658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 11/09/2024] [Accepted: 11/25/2024] [Indexed: 01/16/2025] Open
Abstract
Probiotics are live microorganisms that, when administered in adequate amounts, provide health benefits to the host. According to the International Society of Sports Nutrition (ISSN), probiotic supplementation can optimize the health, performance, and recovery of athletes at all stages of their careers. Recent research suggests that probiotics can improve immune system functions, reduce gastrointestinal distress, and increase gut permeability in athletes. Additionally, probiotics may provide athletes with secondary health benefits that could positively affect athletic performance through enhanced recovery from fatigue, improved immune function, and maintenance of healthy gastrointestinal tract function. The integration of some probiotic strains into athletes' diets and the consumption of multi-strain compounds may lead to an improvement in performance and can positively affect performance-related aspects such as fatigue, muscle pain, body composition, and cardiorespiratory fitness. In summary, probiotics can be beneficial for athletes at all stages of their careers, from amateur to professional. This paper reviews the progress of research on the role of probiotic supplementation in improving energy metabolism and immune system functions, reducing gastrointestinal distress, and enhancing recovery from fatigue in athletes at different levels.
Collapse
Affiliation(s)
- Yousef Nami
- Department of Food Biotechnology, Branch for Northwest and West RegionAgricultural Biotechnology Research Institute of Iran, Agricultural Research, Education and Extension Organization (AREEO)TabrizIran
| | - Anahita Barghi
- Institute of Agricultural Life ScienceDong‐A UniversityBusanSouth Korea
| | - Mehdi Shahgolzari
- Department of Medical Nanotechnology, Faculty of Advanced Medical SciencesTabriz University of Medical SciencesTabrizIran
- Biotechnology Research CenterTabriz University of Medical SciencesTabrizIran
| | - Melika Salehian
- Student Research CommitteeKermanshah University of Medical SciencesKermanshahIran
| | - Babak Haghshenas
- Regenerative Medicine Research Center (RMRC), Health Technology InstituteKermanshah University of Medical SciencesKermanshahIran
| |
Collapse
|
8
|
Kowalczyk M, Radziwill-Bienkowska JM, Marć MA, Jastrząb R, Mytych J, Siedlecki P, Szczepankowska AK. Screening for probiotic properties and potential immunogenic effects of lactobacilli strains isolated from various food products. Front Microbiol 2024; 15:1430582. [PMID: 39534506 PMCID: PMC11554495 DOI: 10.3389/fmicb.2024.1430582] [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: 05/10/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Deceleration of disease progression and re-establishment of microbial balance in the gut is often achieved by application of lactobacilli strains. Their beneficial effects are associated with probiotic properties, which may be accompanied by immunomodulatory action at mucosal surfaces. Methods To single out such strains, we screened almost three hundred lactobacilli isolates from eight genera and various food sources for acid and bile salt tolerance, adhesion to mucin as well as hemolytic activity and antibiotic susceptibility. Moreover, the immunomodulatory effects of cell-free supernatant (CFS) fractions of five lactobacilli strains were assessed using an in vitro cell line model. Results and discussion By our rationalized selection approach, we identified thirty-five strains with probiotic potential and biosafety features. Additionally, we showed that CFS from the Lactiplantibacillus L_4 strain downregulates proinflammatory cytokines IL-8 and IL-1β. In contrast, IL-8 expression was found to increase after treatment with CFSs of Lactiplantibacillus L_2 and L_5 and IL-1β was upregulated by CFSs of Lactiplantibacillus L_1 and Lactiplantibacillus L_3. Overall, our result delineate a rational approach of selecting lactobacilli strains for probiotic development to support the gut microbiota equilibrium and reinforce the host immune system.
Collapse
Affiliation(s)
- Magdalena Kowalczyk
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | | | | - Rafał Jastrząb
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
- Research and Development Center, Olimp Laboratories, Dębica, Poland
| | - Jennifer Mytych
- Research and Development Center, Olimp Laboratories, Dębica, Poland
| | - Paweł Siedlecki
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | | |
Collapse
|
9
|
Barone M, Martucci M, Sciara G, Conte M, Medina LSJ, Iattoni L, Miele F, Fonti C, Franceschi C, Brigidi P, Salvioli S, Provini F, Turroni S, Santoro A. Towards a personalized prediction, prevention and therapy of insomnia: gut microbiota profile can discriminate between paradoxical and objective insomnia in post-menopausal women. EPMA J 2024; 15:471-489. [PMID: 39239112 PMCID: PMC11371979 DOI: 10.1007/s13167-024-00369-1] [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: 02/26/2024] [Accepted: 05/23/2024] [Indexed: 09/07/2024]
Abstract
Background Insomnia persists as a prevalent sleep disorder among middle-aged and older adults, significantly impacting quality of life and increasing susceptibility to age-related diseases. It is classified into objective insomnia (O-IN) and paradoxical insomnia (P-IN), where subjective and objective sleep assessments diverge. Current treatment regimens for both patient groups yield unsatisfactory outcomes. Consequently, investigating the neurophysiological distinctions between P-IN and O-IN is imperative for devising novel precision interventions aligned with primary prediction, targeted prevention, and personalized medicine (PPPM) principles.Working hypothesis and methodology.Given the emerging influence of gut microbiota (GM) on sleep physiology via the gut-brain axis, our study focused on characterizing the GM profiles of a well-characterized cohort of 96 Italian postmenopausal women, comprising 54 insomniac patients (18 O-IN and 36 P-IN) and 42 controls, through 16S rRNA amplicon sequencing. Associations were explored with general and clinical history, sleep patterns, stress, hematobiochemical parameters, and nutritional patterns. Results Distinctive GM profiles were unveiled between O-IN and P-IN patients. O-IN patients exhibited prominence in the Coriobacteriaceae family, including Collinsella and Adlercreutzia, along with Erysipelotrichaceae, Clostridium, and Pediococcus. Conversely, P-IN patients were mainly discriminated by Bacteroides, Staphylococcus, Carnobacterium, Pseudomonas, and respective families, along with Odoribacter. Conclusions These findings provide valuable insights into the microbiota-mediated mechanism of O-IN versus P-IN onset. GM profiling may thus serve as a tailored stratification criterion, enabling the identification of women at risk for specific insomnia subtypes and facilitating the development of integrated microbiota-based predictive diagnostics, targeted prevention, and personalized therapies, ultimately enhancing clinical effectiveness. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00369-1.
Collapse
Affiliation(s)
- Monica Barone
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Institute of Information Technologies, Mathematics and Mechanics, and Institute of Biogerontology, Lobachevsky State University, Nizhny Novgorod, Russia
- IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Interdepartmental Centre "Alma Mater Research Institute On Global Challenges and Climate Change (Alma Climate)", University of Bologna, Bologna, Italy
| | - Morena Martucci
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Institute of Information Technologies, Mathematics and Mechanics, and Institute of Biogerontology, Lobachevsky State University, Nizhny Novgorod, Russia
- IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
- Interdepartmental Centre "Alma Mater Research Institute On Global Challenges and Climate Change (Alma Climate)", University of Bologna, Bologna, Italy
| | - Giuseppe Sciara
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Maria Conte
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Lorenzo Iattoni
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Filomena Miele
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Cristina Fonti
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Claudio Franceschi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Institute of Information Technologies, Mathematics and Mechanics, and Institute of Biogerontology, Lobachevsky State University, Nizhny Novgorod, Russia
| | - Patrizia Brigidi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Stefano Salvioli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | - Federica Provini
- IRCCS Istituto Delle Scienze Neurologiche Di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Aurelia Santoro
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Interdepartmental Centre "Alma Mater Research Institute On Global Challenges and Climate Change (Alma Climate)", University of Bologna, Bologna, Italy
| |
Collapse
|
10
|
Dhakephalkar T, Pisu V, Margale P, Chandras S, Shetty D, Wagh S, Dagar SS, Kapse N, Dhakephalkar PK. Strain-Dependent Adhesion Variations of Shouchella clausii Isolated from Healthy Human Volunteers: A Study on Cell Surface Properties and Potential Probiotic Benefits. Microorganisms 2024; 12:1771. [PMID: 39338446 PMCID: PMC11434523 DOI: 10.3390/microorganisms12091771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/30/2024] Open
Abstract
The probiotic potential of Shouchella clausii is widely recognized, but little is known about its adhesive properties. Hence, this study aims to investigate the adhesion potential and cell surface properties of four human-origin S. clausii strains (B619/R, B603/Nb, B106, and B637/Nm). We evaluated epithelial adhesion, Extracellular Matrix (ECM) binding, aggregation ability, and cell surface hydrophobicity and used genome analysis for validation. Our results demonstrate that adhesion capability is a strain-specific attribute, with significant variations observed among the four strains. B619/R, B603/Nb, and B106 displayed stronger adhesion properties than B637/Nm. Supplementary adhesion assays showed that B637/Nm displayed high hydrophobicity, significant auto-aggregation, and significant mucin-binding abilities. Conversely, B619/R, B603/Nb, and B106 had mildly hydrophobic surfaces and low aggregation abilities. Genome annotation revealed the presence of various adhesion proteins in four strains. Notably, the reduced adhesion potential of B637/Nm was supported by the absence of the cell wall surface anchor family protein (LPxTG motif), which is crucial for interactions with intestinal epithelial cells or mucus components. Further, docking studies provided insights into the interaction of adhesion proteins with gut mucins. These findings contribute to a better understanding of how S. clausii strains interact with the gut environment, facilitating the development of probiotic formulations tailored for improved gut health and well-being.
Collapse
Affiliation(s)
- Tanisha Dhakephalkar
- Hi Tech BioSciences India Ltd., Research & Development Centre, Plot No. 6 and 8, Ambadvet Industrial Estate, PO Paud, Pune 412108, Maharashtra, India
| | - Vaidehi Pisu
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| | - Prajakta Margale
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
| | - Siddhi Chandras
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| | - Deepa Shetty
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
| | - Shilpa Wagh
- Hi Tech BioSciences India Ltd., Research & Development Centre, Plot No. 6 and 8, Ambadvet Industrial Estate, PO Paud, Pune 412108, Maharashtra, India
| | - Sumit Singh Dagar
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| | - Neelam Kapse
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
| | - Prashant K Dhakephalkar
- Bioenergy Group, MACS-Agharkar Research Institute, G.G. Agarkar Road, Pune 411004, Maharashtra, India
- Department of Microbiology, Savitribai Phule Pune University, Ganeshkhind Rd., Aundh, Pune 411007, Maharashtra, India
| |
Collapse
|
11
|
Kozawa T, Aoyagi H. Novel method for screening probiotic candidates tolerant to human gastrointestinal stress. J Microbiol Methods 2024; 222:106945. [PMID: 38729266 DOI: 10.1016/j.mimet.2024.106945] [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: 03/22/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/12/2024]
Abstract
Tolerance to human gastrointestinal stressors is crucial for probiotics to exhibit their health benefits; however, there is no standardised method for screening their stress tolerance. In this study, we proposed a novel method for screening probiotic candidates tolerant to human gastrointestinal stress-gastrointestinal tolerance assay and culture (GTA-C) method-using black polyethylene terephthalate (PET) non-woven fabric as a scaffold to modify the specialized cellulose film (SCF) method. The modified SCF method showed excellent pH-based diffusion of medium components, had minimal effect on the growth of Escherichia coli K12, and improved the visibility of the colonies. Analysis of kimchi samples cultured using the SCF and modified SCF methods revealed that the modified method diversified the cultured bacteria. GTA in a simulated human fasting state using the modified SCF method showed that acid stress significantly affected the growth of four bacteria used as probiotics and that tolerance to acid stress may be species-dependent. Screening of probiotics in kimchi samples resulted in the identification of lactic acid bacteria tolerant to human gastrointestinal stress during fasting. Our results indicate that the modified SCF method (GTA-C method) is useful for screening probiotics resistant to the gastrointestinal environment during fasting.
Collapse
Affiliation(s)
- Takuma Kozawa
- Master's Program in Agro-Bioresources Science and Technology, Degree Programs in Life and Earth Sciences, Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| | - Hideki Aoyagi
- Institute of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan; Microbiology Research Center for Sustainability (MiCS), University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan.
| |
Collapse
|
12
|
Ghannoum MA, Elshaer M, Al-Shakhshir H, Retuerto M, McCormick TS. A Probiotic Amylase Blend Positively Impacts Gut Microbiota Modulation in a Randomized, Placebo-Controlled, Double-Blind Study. Life (Basel) 2024; 14:824. [PMID: 39063578 PMCID: PMC11277872 DOI: 10.3390/life14070824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The present study was performed to determine if ingesting a blend of probiotics plus amylase would alter the abundance and diversity of gut microbiota in subjects consuming the blend over a 6-week period. 16S and ITS ribosomal RNA (rRNA) sequencing was performed on fecal samples provided by subjects who participated in a clinical study where they consumed either a probiotic amylase blend (Bifidobacterium breve 19bx, Lactobacillus acidophilus 16axg, Lacticaseibacillus rhamnosus 18fx, and Saccharomyces boulardii 16mxg, alpha amylase (500 SKB (Alpha-amylase-Dextrinizing Units)) or a placebo consisting of rice oligodextrin. The abundance and diversity of both bacterial and fungal organisms was assessed at baseline and following 6 weeks of probiotic amylase blend or placebo consumption. In the subjects consuming the probiotic blend, the abundance of Saccharomyces cerevisiae increased 200-fold, and its prevalence increased (~20% to ~60%) (p ≤ 0.05), whereas the potential pathogens Bacillus thuringiensis and Macrococcus caseolyticus decreased more than 150- and 175-fold, respectively, after probiotic-amylase blend consumption. We also evaluated the correlation between change in microbiota and clinical features reported following probiotic amylase consumption. Nine (9) species (seven bacterial and two fungal) were significantly (negatively or positively) associated with the change in 32 clinical features that were originally evaluated in the clinical study. Oral supplementation with the probiotic-amylase blend caused a marked increase in abundance of the beneficial yeast S. cerevisiae and concomitant modulation of gut-dwelling commensal bacterial organisms, providing the proof of concept that a beneficial commensal organism can re-align the gut microbiota.
Collapse
Affiliation(s)
- Mahmoud A. Ghannoum
- Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| | - Mohammed Elshaer
- Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
- Department of Clinical Pathology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Hilmi Al-Shakhshir
- Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Mauricio Retuerto
- Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Thomas S. McCormick
- Department of Dermatology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
| |
Collapse
|
13
|
Bermúdez-Humarán LG, Chassaing B, Langella P. Exploring the interaction and impact of probiotic and commensal bacteria on vitamins, minerals and short chain fatty acids metabolism. Microb Cell Fact 2024; 23:172. [PMID: 38867272 PMCID: PMC11167913 DOI: 10.1186/s12934-024-02449-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 06/04/2024] [Indexed: 06/14/2024] Open
Abstract
There is increasing evidence that probiotic and commensal bacteria play a role in substrate metabolism, energy harvesting and intestinal homeostasis, and may exert immunomodulatory activities on human health. In addition, recent research suggests that these microorganisms interact with vitamins and minerals, promoting intestinal and metabolic well-being while producing vital microbial metabolites such as short-chain fatty acids (SCFAs). In this regard, there is a flourishing field exploring the intricate dynamics between vitamins, minerals, SCFAs, and commensal/probiotic interactions. In this review, we summarize some of the major hypotheses beyond the mechanisms by which commensals/probiotics impact gut health and their additional effects on the absorption and metabolism of vitamins, minerals, and SCFAs. Our analysis includes comprehensive review of existing evidence from preclinical and clinical studies, with particular focus on the potential interaction between commensals/probiotics and micronutrients. Finally, we highlight knowledge gaps and outline directions for future research in this evolving field.
Collapse
Affiliation(s)
- Luis G Bermúdez-Humarán
- Laboratory of Commensals and Probiotics-Host Interactions, Université Paris-Saclay, INRAE, Micalis Institute, Jouy-en-Josas, AgroParisTech, 78350, France.
| | - Benoit Chassaing
- Microbiome-Host Interactions, Institut Pasteur, Université Paris Cité, INSERM U1306, Paris, France
- INSERM U1016, team Mucosal microbiota in chronic inflammatory diseases, CNRS UMR 8104, Université de Paris, Paris, France
| | - Philippe Langella
- Laboratory of Commensals and Probiotics-Host Interactions, Université Paris-Saclay, INRAE, Micalis Institute, Jouy-en-Josas, AgroParisTech, 78350, France.
| |
Collapse
|
14
|
Hou L, Fu Y, Zhao C, Fan L, Hu H, Yin S. The research progress on the impact of antibiotics on the male reproductive system. ENVIRONMENT INTERNATIONAL 2024; 187:108670. [PMID: 38669720 DOI: 10.1016/j.envint.2024.108670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024]
Abstract
Antibiotics are extensively utilized in the livestock and poultry industry and can accumulate in animals and the environment, leading to potential health risks for humans via food and water consumption. Research on antibiotic toxicity, particularly their impact as endocrine disruptors on the male reproductive system, is still in its nascent stages. This review highlights the toxic effect of antibiotics on the male reproductive system, detailing the common routes of exposure and the detrimental impact and mechanisms of various antibiotic classes. Additionally, it discusses the protective role of food-derived active substances against the reproductive toxicity induced by antibiotics. This review aims to raise awareness about the reproductive toxicity of antibiotics in males and to outline the challenges that must be addressed in future research.
Collapse
Affiliation(s)
- Lirui Hou
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Yuhan Fu
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Chong Zhao
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Lihong Fan
- College of Veterinary Medicine, China Agricultural University, Yunamingyuan West Road, Haidian District, Beijing 100193, China
| | - Hongbo Hu
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua East Road, Haidian District, Beijing 100083, China
| | - Shutao Yin
- Department of Nutrition and Health, College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua East Road, Haidian District, Beijing 100083, China.
| |
Collapse
|
15
|
Holcomb M, Marshall A, Flinn H, Lozano M, Soriano S, Gomez-Pinilla F, Treangen TJ, Villapol S. Probiotic treatment causes sex-specific neuroprotection after traumatic brain injury in mice. RESEARCH SQUARE 2024:rs.3.rs-4196801. [PMID: 38645104 PMCID: PMC11030542 DOI: 10.21203/rs.3.rs-4196801/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Background Recent studies have shed light on the potential role of gut dysbiosis in shaping traumatic brain injury (TBI) outcomes. Changes in the levels and types of Lactobacillus bacteria present might impact the immune system disturbances, neuroinflammatory responses, anxiety and depressive-like behaviors, and compromised neuroprotection mechanisms triggered by TBI. Objective This study aimed to investigate the effects of a daily pan-probiotic (PP) mixture in drinking water containing strains of Lactobacillus plantarum, L. reuteri, L. helveticus, L. fermentum, L. rhamnosus, L. gasseri, and L. casei, administered for either two or seven weeks before inducing TBI on both male and female mice. Methods Mice were subjected to controlled cortical impact (CCI) injury. Short-chain fatty acids (SCFAs) analysis was performed for metabolite measurements. The taxonomic profiles of murine fecal samples were evaluated using 16S rRNA V1-V3 sequencing analysis. Histological analyses were used to assess neuroinflammation and gut changes post-TBI, while behavioral tests were conducted to evaluate sensorimotor and cognitive functions. Results Our findings suggest that PP administration modulates the diversity and composition of the microbiome and increases the levels of SCFAs in a sex-dependent manner. We also observed a reduction of lesion volume, cell death, and microglial and macrophage activation after PP treatment following TBI in male mice. Furthermore, PP-treated mice show motor function improvements and decreases in anxiety and depressive-like behaviors. Conclusion Our findings suggest that PP administration can mitigate neuroinflammation and ameliorate motor and anxiety and depressive-like behavior deficits following TBI. These results underscore the potential of probiotic interventions as a viable therapeutic strategy to address TBI-induced impairments, emphasizing the need for gender-specific treatment approaches.
Collapse
|
16
|
de Luna Freire MO, Cruz Neto JPR, de Albuquerque Lemos DE, de Albuquerque TMR, Garcia EF, de Souza EL, de Brito Alves JL. Limosilactobacillus fermentum Strains as Novel Probiotic Candidates to Promote Host Health Benefits and Development of Biotherapeutics: A Comprehensive Review. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10235-1. [PMID: 38393628 DOI: 10.1007/s12602-024-10235-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2024] [Indexed: 02/25/2024]
Abstract
Fruits and their processing by-products are sources of potentially probiotic strains. Limosilactobacillus (L.) fermentum strains isolated from fruit processing by-products have shown probiotic-related properties. This review presents and discusses the results of the available studies that evaluated the probiotic properties of L. fermentum in promoting host health benefits, their application by the food industry, and the development of biotherapeutics. The results showed that administration of L. fermentum for 4 to 8 weeks promoted host health benefits in rats, including the modulation of gut microbiota, improvement of metabolic parameters, and antihypertensive, antioxidant, and anti-inflammatory effects. The results also showed the relevance of L. fermentum strains for application in the food industry and for the formulation of novel biotherapeutics, especially nutraceuticals. This review provides evidence that L. fermentum strains isolated from fruit processing by-products have great potential for promoting host health and indicate the need for a translational approach to confirm their effects in humans using randomized, double-blind, placebo-controlled trials.
Collapse
Affiliation(s)
- Micaelle Oliveira de Luna Freire
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I-Jd. Cidade Universitária, João Pessoa, PB, 58051-900, Brazil
| | - José Patrocínio Ribeiro Cruz Neto
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I-Jd. Cidade Universitária, João Pessoa, PB, 58051-900, Brazil
| | | | | | - Estefânia Fernandes Garcia
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I-Jd. Cidade Universitária, João Pessoa, PB, 58051-900, Brazil
| | - Evandro Leite de Souza
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I-Jd. Cidade Universitária, João Pessoa, PB, 58051-900, Brazil
| | - José Luiz de Brito Alves
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, Campus I-Jd. Cidade Universitária, João Pessoa, PB, 58051-900, Brazil.
| |
Collapse
|
17
|
Paterniti I, Scuderi SA, Cambria L, Nostro A, Esposito E, Marino A. Protective Effect of Probiotics against Pseudomonas aeruginosa Infection of Human Corneal Epithelial Cells. Int J Mol Sci 2024; 25:1770. [PMID: 38339047 PMCID: PMC10855269 DOI: 10.3390/ijms25031770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Probiotic therapy needs consideration as an alternative strategy to prevent and possibly treat corneal infection. This study aimed to assess the preventive effect of Lactobacillus reuteri and Bifidobacterium longum subsp. infantis on reducing the infection of human corneal epithelial (HCE) cells caused by Pseudomonas aeruginosa. The probiotics' preventive effect against infection was evaluated in cell monolayers pretreated with each probiotic 1 h and 24 h prior to P. aeruginosa challenge followed by 1 h and 24 h of growth in combination. Cell adhesion, cytotoxicity, anti-inflammatory, and antinitrosative activities were evaluated. L. reuteri and B. longum adhered to HCE cells, preserved occludin tight junctions' integrity, and increased mucin production on a SkinEthicTM HCE model. Pretreatment with L. reuteri or B. longum significantly protected HCE cells from infection at 24 h, increasing cell viability at 110% (110.51 ± 5.15; p ≤ 0.05) and 137% (137.55 ± 11.97; p ≤ 0.05), respectively. Each probiotic showed anti-inflammatory and antinitrosative activities, reducing TNF-α level (p ≤ 0.001) and NOx amount (p ≤ 0.001) and reestablishing IL-10 level (p ≤ 0.001). In conclusion, this study demonstrated that L. reuteri and B. longum exert protective effects in the context of corneal infection caused by P. aeruginosa by restoring cell viability and modulating inflammatory cytokine release.
Collapse
Affiliation(s)
| | | | | | | | | | - Andreana Marino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98166 Messina, Italy; (I.P.); (S.A.S.); (L.C.); (A.N.); (E.E.)
| |
Collapse
|
18
|
Guo D, Deng Y, Yang Q, Li M, Wang X, Wan X, He J, Xu Y, Huang W, Lin G, Xu Y, Sun Y, Zhang R, Chen WH, Liu Z. Symbiotic probiotic communities with multiple targets successfully combat obesity in high-fat-diet-fed mice. Gut Microbes 2024; 16:2420771. [PMID: 39488738 PMCID: PMC11540072 DOI: 10.1080/19490976.2024.2420771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/24/2024] [Accepted: 10/19/2024] [Indexed: 11/04/2024] Open
Abstract
Probiotics hold great potential for treating metabolic diseases such as obesity. Given the complex and multifactorial nature of these diseases, research on probiotic combination with multiple targets has become popular. Here, we choose four obesity-related targets to perform high-throughput screening, including pancreatic lipase activity, bile salt hydrolase activity, glucagon-like peptide-1 secretion and adipocyte differentiation. Then, we obtained 649 multi-strain combinations with the requirement that each must cover all these targets in principle. After in vitro co-culture and in vivo co-colonization experiments, only four (<0.7%) combinations were selected as symbiotic probiotic communities (SPCs). Next, genome-scale metabolic model analysis revealed that these SPCs showed lower metabolic resource overlap and higher metabolic interaction potential involving amino acid metabolism (Ammonium, L-Lysine, etc.) and energy metabolism (Phosphate, etc.). Further animal experiments demonstrated that all SPCs exhibited a good safety profile and excellent effects in improving obesity and associated glucose metabolism disruptions and depression-like behaviors in high-fat-diet-fed mice. This anti-obesity improvement was achieved through reduced cholesterol level, fat accumulation and inhibited adipocyte differentiation. Taken together, our study provides a new perspective for designing multi-strain combinations, which may facilitate greater therapeutic effect on obesity and other complex diseases in the future.
Collapse
Affiliation(s)
- Dingming Guo
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yun Deng
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qianqian Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Min Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiangfeng Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuchun Wan
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junqing He
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ying Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Wenxin Huang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Guohua Lin
- Biotechnology Department, Biological Anti-Aging Academy of Wuhan East Lake High-tech Development Zone,Wuhan,China
| | - Ya Xu
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Yi Sun
- Department of Clinical Laboratory, The First People’s Hospital of Yunnan Province,The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Ruilin Zhang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, China
| | - Wei-Hua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhi Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
19
|
Jaafar MH, Xu P, Mageswaran UM, Balasubramaniam SD, Solayappan M, Woon JJ, Teh CSJ, Todorov SD, Park YH, Liu G, Liong MT. Constipation anti-aging effects by dairy-based lactic acid bacteria. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2024; 66:178-203. [PMID: 38618031 PMCID: PMC11007456 DOI: 10.5187/jast.2023.e93] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/24/2023] [Accepted: 09/01/2023] [Indexed: 04/16/2024]
Abstract
Constipation, which refers to difficulties in defecation and infrequent bowel movement in emptying the gastrointestinal system that ultimately produces hardened fecal matters, is a health concern in livestock and aging animals. The present study aimed to evaluate the potential effects of dairy-isolated lactic acid bacteria (LAB) strains to alleviate constipation as an alternative therapeutic intervention for constipation treatment in the aging model. Rats were aged via daily subcutaneous injection of D-galactose (600 mg/body weight [kg]), prior to induction of constipation via oral administration of loperamide hydrochloride (5 mg/body weight [kg]). LAB strains (L. fermentum USM 4189 or L. plantarum USM 4187) were administered daily via oral gavage (1 × 10 Log CFU/day) while the control group received sterile saline. Aged rats as shown with shorter telomere lengths exhibited increased fecal bulk and soften fecal upon administration of LAB strains amid constipation as observed using the Bristol Stool Chart, accompanied by a higher fecal moisture content as compared to the control (p < 0.05). Fecal water-soluble metabolite profiles showed a reduced concentration of threonine upon administration of LAB strains compared to the control (p < 0.05). Histopathological analysis also showed that the administration of LAB strains contributed to a higher colonic goblet cell count as compared to the control (p < 0.05). The present study illustrates the potential of dairy-sourced LAB strains as probiotics to ameliorate the adverse effect of constipation amid aging, and as a potential dietary intervention strategy for dairy foods including yogurt and cheese.
Collapse
Affiliation(s)
- Mohamad Hafis Jaafar
- Bioprocess Technology, School of
Industrial Technology, Universiti Sains Malaysia, Penang
11800, Malaysia
| | - Pei Xu
- Bioprocess Technology, School of
Industrial Technology, Universiti Sains Malaysia, Penang
11800, Malaysia
- Faculty of Cuisine, Sichuan Tourism
University, Chengdu 610100, China
| | - Uma-Mageswary Mageswaran
- Bioprocess Technology, School of
Industrial Technology, Universiti Sains Malaysia, Penang
11800, Malaysia
| | | | | | - Jia-Jie Woon
- Department of Medical Microbiology,
Faculty of Medicine, University of Malaya, Kuala Lumpur 50603,
Malaysia
| | - Cindy Shuan-Ju Teh
- Department of Medical Microbiology,
Faculty of Medicine, University of Malaya, Kuala Lumpur 50603,
Malaysia
| | - Svetoslav Dimitrov Todorov
- ProBacLab, Department of Food Science and
Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of
Sao Paulo, Sao Paulo 05508-090, Brazil
| | | | - Guoxia Liu
- CAS Key Laboratory of Microbial
Physiological and Metabolic Engineering, State Key Laboratory of Microbial
Resources, Institute of Microbiology, Chinese Academy of
Sciences, Beijing 100864, China
- CAS-TWAS Centre of Excellence for
Biotechnology, Beijing 100101, China
| | - Min-Tze Liong
- Bioprocess Technology, School of
Industrial Technology, Universiti Sains Malaysia, Penang
11800, Malaysia
- Renewable Biomass Transformation
Cluster, School of Industrial Technology, Universiti Sains
Malaysia, Penang 11800, Malaysia
| |
Collapse
|
20
|
Sun XW, Huang HJ, Wang XM, Wei RQ, Niu HY, Chen HY, Luo M, Abdugheni R, Wang YL, Liu FL, Jiang H, Liu C, Liu SJ. Christensenella strain resources, genomic/metabolomic profiling, and association with host at species level. Gut Microbes 2024; 16:2347725. [PMID: 38722028 PMCID: PMC11085954 DOI: 10.1080/19490976.2024.2347725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 03/15/2024] [Accepted: 04/22/2024] [Indexed: 05/12/2024] Open
Abstract
The gut commensal bacteria Christensenellaceae species are negatively associated with many metabolic diseases, and have been seen as promising next-generation probiotics. However, the cultured Christensenellaceae strain resources were limited, and their beneficial mechanisms for improving metabolic diseases have yet to be explored. In this study, we developed a method that enabled the enrichment and cultivation of Christensenellaceae strains from fecal samples. Using this method, a collection of Christensenellaceae Gut Microbial Biobank (ChrisGMB) was established, composed of 87 strains and genomes that represent 14 species of 8 genera. Seven species were first described and the cultured Christensenellaceae resources have been significantly expanded at species and strain levels. Christensenella strains exerted different abilities in utilization of various complex polysaccharides and other carbon sources, exhibited host-adaptation capabilities such as acid tolerance and bile tolerance, produced a wide range of volatile probiotic metabolites and secondary bile acids. Cohort analyses demonstrated that Christensenellaceae and Christensenella were prevalent in various cohorts and the abundances were significantly reduced in T2D and OB cohorts. At species level, Christensenellaceae showed different changes among healthy and disease cohorts. C. faecalis, F. tenuis, L. tenuis, and Guo. tenuis significantly reduced in all the metabolic disease cohorts. The relative abundances of C. minuta, C. hongkongensis and C. massiliensis showed no significant change in NAFLD and ACVD. and C. tenuis and C. acetigenes showed no significant change in ACVD, and Q. tenuis and Geh. tenuis showed no significant change in NAFLD, when compared with the HC cohort. So far as we know, this is the largest collection of cultured resource and first exploration of Christensenellaceae prevalences and abundances at species level.
Collapse
Affiliation(s)
- Xin-Wei Sun
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Hao-Jie Huang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Xiao-Meng Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Rui-Qi Wei
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Han-Yu Niu
- College of Veterinary Medicine, Shanxi Agr icultural University, Taigu, China
| | - Hao-Yu Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Man Luo
- College of Veterinary Medicine, Shanxi Agr icultural University, Taigu, China
| | - Rashidin Abdugheni
- State Key Laboratory of Desert and Oasis Ecology, Key Laboratory of Ecological Safety and Sustainable Development in Arid Lands, Xinjiang Institute of Ecology and Geography, Chinese Academy of Sciences, Ürűmqi, P. R. China
| | - Yu-Lin Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Feng-Lan Liu
- College of Life Sciences, Hebei University, Baoding, P. R. China
| | - He Jiang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Chang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
| | - Shuang-Jiang Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, P. R. China
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, P. R. China
| |
Collapse
|
21
|
Rathi A, Pagare R. Efficacy and Safety of Bacillus coagulans LBSC in Drug Induced Constipation Associated With Functional Gastrointestinal Disorder: A Double-Blind, Randomized, Interventional, Parallel, Controlled Trial a Clinical Study on Bacillus coagulans LBSC for Drug Induced Constipation Associated With FGIDs. GLOBAL ADVANCES IN INTEGRATIVE MEDICINE AND HEALTH 2024; 13:27536130241286511. [PMID: 39295947 PMCID: PMC11409293 DOI: 10.1177/27536130241286511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 09/02/2024] [Accepted: 09/08/2024] [Indexed: 09/21/2024]
Abstract
Background Active drugs and nutraceutical supplements commonly induce various gastrointestinal illnesses, and constipation is a major gastrointestinal symptom accompanied with functional gastrointestinal disorders. Drug-induced imbalance in gut microbiota may play critical role in such physiological disturbances. Probiotics have been known for resuming normal and healthy gut microbiome. Objective To investigate the clinical efficacy and safety of Bacillus coagulans LBSC in the treatment of drug induced constipation associated with functional gastrointestinal disorder (FGID) symptoms. Methods A prospective, interventional, randomized, double-blind, parallel, multi-arm, controlled trial with 168 patients experiencing drug induced constipation associated with FGID symptoms (DICAWFGID) screened through Rome IV criteria were randomized into 2 arms, i.e. placebo arm (n = 28) and atorvastatin, atenolol, metformin, amitriptyline, and calcium in test arm (n = 28/arm). Patients in both arms received similar dosages (1 g sachet, 3 times a day) for 35 days. The occurrence of constipation using Bristol Stool Form Scale, assessment of degree of constipation on 4-point Likert scale, occurrence of hard stool and degree of stool expulsion on 3-point scale, and defecation frequency were primary endpoints. While, secondary outcomes consisted of the changes in severity of FGID symptoms, visual analogue scale and tolerance to IP, along with reports of adverse events (AEs) and severe adverse events (SAEs). Results There was a significant reduction in occurrence of constipation (≥98.6% and P-value <0.05) in test arm over the placebo arm. Assessment of co-primary endpoints showed significant improvements in degree of stool consistency (P-value 0.0232; CI: 0.1870, 1.1629), borderline significantly superior in degree of stool expulsion (P-value 0.0553; CI: 0.0378, -0.4939), while the other co-primary efficacy endpoints displayed considerably improved advancement (non-significant, P-value ≥0.05). The intra group analysis of symptoms at start of treatment (SOT) and end of treatment (EOT) revealed a significant reduction in scores for occurrence of constipation and degree of constipation, whereas significant improvement in the scores for degree of stool consistency and degree of stool expulsion (P-value <0.001) after the intervention period. In secondary endpoints, the processed responses clearly signified a considerable positive improvement (non-significant, P-value ≥0.05) in other symptoms of constipation associated with FGIDs as determined by the changes in the EOT-SOT score. The study data also highlighted the safety of Bacillus coagulans LBSC at the studied dose. No AEs and/or SAEs were documented during the investigation. Conclusion At the studied dose, Bacillus coagulans LBSC was safe for oral consumption and effective in the management of the drug induced constipation associated with FGIDs symptoms.
Collapse
Affiliation(s)
- Ankit Rathi
- Department of Biological Sciences, School of Science, Sandip University, Nashik, Maharashtra, India
| | - Ravikiran Pagare
- Department of Biological Sciences, School of Science, Sandip University, Nashik, Maharashtra, India
| |
Collapse
|
22
|
Abuqwider J, Di Porzio A, Barrella V, Gatto C, Sequino G, De Filippis F, Crescenzo R, Spagnuolo MS, Cigliano L, Mauriello G, Iossa S, Mazzoli A. Limosilactobacillus reuteri DSM 17938 reverses gut metabolic dysfunction induced by Western diet in adult rats. Front Nutr 2023; 10:1236417. [PMID: 37908302 PMCID: PMC10613642 DOI: 10.3389/fnut.2023.1236417] [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: 06/07/2023] [Accepted: 09/18/2023] [Indexed: 11/02/2023] Open
Abstract
Introduction Microencapsulation of probiotic bacteria is an efficient and innovative new technique aimed at preserving bacterial survival in the hostile conditions of the gastrointestinal tract. However, understanding whether a microcapsule preserves the effectiveness of the bacterium contained within it is of fundamental importance. Methods Male Wistar rats aged 90 days were fed a control diet or a Western diet for 8 weeks, with rats fed the Western diet divided into three groups: one receiving the diet only (W), the second group receiving the Western diet and free L. reuteri DSM 17938 (WR), and the third group receiving the Western diet and microencapsulated L. reuteri DSM 17938 (WRM). After 8 weeks of treatment, gut microbiota composition was evaluated, together with occludin, one of the tight junction proteins, in the ileum and the colon. Markers of inflammation were also quantified in the portal plasma, ileum, and colon, as well as markers for gut redox homeostasis. Results The Western diet negatively influenced the intestinal microbiota, with no significant effect caused by supplementation with free and microencapsulated L. reuteri. However, L. reuteri, in both forms, effectively preserved the integrity of the intestinal barrier, thus protecting enterocytes from the development of inflammation and oxidative stress. Conclusion From these whole data, it emerges that L. reuteri DSM 17938 can be an effective probiotic in preventing the unhealthy consequences of the Western diet, especially in the gut, and that microencapsulation preserves the probiotic effects, thus opening the formulation of new preparations to be able to improve gut function independent of dietary habits.
Collapse
Affiliation(s)
- Jumana Abuqwider
- Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy
| | - Angela Di Porzio
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Valentina Barrella
- Department of Biology, University of Naples Federico II, Naples, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
| | - Cristina Gatto
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Giuseppina Sequino
- Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy
| | - Francesca De Filippis
- Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy
| | | | - Maria Stefania Spagnuolo
- Department of Bio-Agrofood Science, Institute for the Animal Production System in the Mediterranean Environment, National Research Council Naples (CNR-ISPAAM), Naples, Italy
| | - Luisa Cigliano
- Department of Biology, University of Naples Federico II, Naples, Italy
| | - Gianluigi Mauriello
- Department of Agricultural Sciences, University of Naples Federico II, Naples, Italy
| | - Susanna Iossa
- Department of Biology, University of Naples Federico II, Naples, Italy
- NBFC, National Biodiversity Future Center, Palermo, Italy
| | - Arianna Mazzoli
- Department of Biology, University of Naples Federico II, Naples, Italy
| |
Collapse
|
23
|
Xi Y, Zhang C, Feng Y, Zhao S, Zhang Y, Duan G, Wang W, Wang J. Genetically predicted the causal relationship between gut microbiota and infertility: bidirectional Mendelian randomization analysis in the framework of predictive, preventive, and personalized medicine. EPMA J 2023; 14:405-416. [PMID: 37605651 PMCID: PMC10439866 DOI: 10.1007/s13167-023-00332-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/26/2023] [Indexed: 08/23/2023]
Abstract
Objective Several studies have reported the association between gut microbiota and infertility; however, the causal association between them remains unclear. This study aimed to explore the causal relationship between gut microbiota and infertility and evaluate how specific gut microbiota can support early monitoring and prevention of infertility in the context of predictive, preventive, and personalized medicine (PPPM/3PM). Methods The gut microbiota GWAS data included 18,340 individuals. Female infertility (6481 cases and 68,969 controls) and male infertility data (680 cases and 72,799 controls) were obtained from the FinnGen consortium. The inverse variance weighting (IVW), MR-Egger, weighted median (WM), Cochran Q tests, MR-PRESSO, and leave-one-out were used as a supplement to Mendelian randomization (MR) results and sensitivity analysis. Results The results of MR analysis indicated a significant causal association between Eubacterium oxidoreducens (OR = 2.048, P = 0.008), Lactococcus (OR = 1.445, P = 0.042), Eubacterium ventriosum (OR = 0.436, P = 0.018), Eubacterium rectale (OR = 0.306, P = 0.002), and Ruminococcaceae NK4A214 (OR = 0.537, P = 0.045) and male infertility. Genetically predicted Eubacterium ventriosum (OR = 0.809, P = 0.018), Holdemania (OR = 0.836, P = 0.037), Lactococcus (OR = 0.867, P = 0.020), Ruminococcaceae NK4A214 (OR = 0.830, P < 0.050), Ruminococcus torques (OR = 0.739, P = 0.022), and Faecalibacterium (OR = 1.311, P = 0.007) were associated with female infertility. Sensitivity analysis did not detect heterogeneity and pleiotropy (P > 0.05). Conclusions Our results provided evidence for the causal relationship between some gut microbiota and male and female infertility. These findings might be valuable in providing personalized treatment options for preventing infertility and improving reproductive function by monitoring and regulating the gut microbiota of infertility patients in the context of PPPM. Moreover, detecting the abundance of microbiota in feces can support preventive and personalized strategies, which may benefit more infertility patients. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-023-00332-6.
Collapse
Affiliation(s)
- Yujia Xi
- Department of Urology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001 Shanxi China
- Second School of Clinical Medicine, Shanxi Medical University, Taiyuan, 030000 China
| | - Chenwei Zhang
- Second School of Clinical Medicine, Shanxi Medical University, Taiyuan, 030000 China
| | - Yiqian Feng
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan, 030000 China
| | - Shurui Zhao
- First School of Clinical Medicine, Shanxi Medical University, Taiyuan, 030000 China
- Department of Obstetrics and Gynecology, The First Hospital Shanxi Medical University, Taiyuan, 030000 China
| | - Yukai Zhang
- School of Basic Medicine, Shanxi Medical University, Taiyuan, 030000 China
| | - Guosheng Duan
- Second School of Clinical Medicine, Shanxi Medical University, Taiyuan, 030000 China
| | - Wei Wang
- Department of Urology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001 Shanxi China
| | - Jingqi Wang
- Department of Urology, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001 Shanxi China
| |
Collapse
|
24
|
Khaleghi M, Khorrami S, Jafari-Nasab T. Pediococcus acidilactici isolated from traditional cheese as a potential probiotic with cytotoxic activity against doxorubicin-resistant MCF-7 cells. 3 Biotech 2023; 13:170. [PMID: 37188290 PMCID: PMC10169992 DOI: 10.1007/s13205-023-03597-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 04/29/2023] [Indexed: 05/17/2023] Open
Abstract
The considerable flexibility of cancerous cells to escape from chemical and biological drugs makes it clear that much is to be done to control and eliminate such cells. Probiotic bacteria, in this regard, have shown promising performance. In this study, we isolated and characterized lactic acid bacteria from traditional cheese. Then we evaluated their activity against doxorubicin-resistant MCF-7 cells (MCF-7/DOX) through MTT assay, Annexin V/PI protocol, real-time PCR, and western blotting. Among the isolates, one strain with more than 97% similarity with Pediococcus acidilactici showed considerable probiotics properties. Low pH, high bile salts, and NaCl could not significantly affect this strain while it was susceptible to antibiotics. Also, it had a potent antibacterial activity. Besides, the cell-free supernatant of this strain (CFS) significantly reduced the viability of MCF-7 and MCF-7/DOX cancerous cells (to about 10% and 25%, respectively), while it was safe for normal cells. Also, we found that CFS could regulate the Bax/Bcl-2 at mRNA and protein levels to induce apoptosis in drug-resistant cells. We determined 75% early apoptosis, 10% late apoptosis, and 15% necrosis in the cells treated with the CFS. These findings can accelerate the development of probiotics as promising alternative treatments to overcome drug-resistant cancers.
Collapse
Affiliation(s)
- Moj Khaleghi
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Sadegh Khorrami
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Tayebeh Jafari-Nasab
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| |
Collapse
|
25
|
Rudyk M, Hurmach Y, Serhiichuk T, Akulenko I, Skivka L, Berehova T, Ostapchenko L. Multi-probiotic consumption sex-dependently interferes with MSG-induced obesity and concomitant phagocyte pro-inflammatory polarization in rats: Food for thought about personalized nutrition. Heliyon 2023; 9:e13381. [PMID: 36816299 PMCID: PMC9932736 DOI: 10.1016/j.heliyon.2023.e13381] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 01/26/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Epidemic scope which obesity has reached in many countries necessitates shifting the emphasis in medicine from traditional reaction to individualized and personalized prevention. Numerous trials convincingly prove sexual dimorphism of obesity in morbidity, pathophysiology, comorbidity, outcomes and prophylaxis efficacy. Obesity is characterized by chronic systemic low-grade inflammation that creates the preconditions for the emergence of numerous comorbidities. Leading role in the initiation, propagation and resolution of inflammation belongs to tissue resident and circulating phagocytes. The outcome of inflammation largely depends on phagocyte functional polarization, which in turn is governed by environmental stimuli. Gut microbiota (GM), whose disturbances are one of the key pathogenetic features in obesity, substantially affect phagocyte functions and can either aggravate or calm obesity-associated inflammation. Probiotics possess promising physiological functions, including microbiota-restoring and anti-inflammatory traits, that may possibly help prevent obesity. However, sex-specific effects of probiotic supplementation for targeted obesity prevention remain unknown. The aim of the current study was aimed to compare the effect of multi-probiotic preparation used in prophylactic regimen on the adiposity, profile of culturable GM and its short-chain fatty acids as well as on functional profile of phagocytes from different locations in male and female rats with monosodium glutamate (MSG)-induced obesity. Obesity was induced by neonatal MSG injections in male and female rats, who were given the multi-species probiotic during juvenile and adult developmental stages. Culturable fecal and mucosa-associated microbiota of the intestine were examined using selective diagnostic media. Short-chain fatty acid profile in fecal samples was determined by GC-MS. Phagocyte functional profile was evaluated using flow cytometry and colorimetric methods. Probiotic supplementation after the administration of MSG prevented weight gain and fat accumulation, inflammatory phagocyte activation and alterations in GM in female rats. In male MSG-injected rats, probiotic supplementation restricted but did not prevent weight gain and fat deposition, alleviated but did not prevent systemic inflammation, prevented the alterations in GM, but with residual imbalance in the ratio of obligate anaerobic to facultative anaerobic bacteria. Our findings emphasize the necessity of sex-centered approaches to the prophylactic use of probiotics in obesity in the context of predictive preventive and personalized medicine.
Collapse
Affiliation(s)
- Mariia Rudyk
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine,Corresponding author.
| | - Yevheniia Hurmach
- Bogomolets National Medical University, 13, T. Shevchenko Blvd, Kyiv, 01601, Ukraine
| | - Tetiana Serhiichuk
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Iryna Akulenko
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Larysa Skivka
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Tetiana Berehova
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| | - Liudmyla Ostapchenko
- Educational and Scientific Centre “Institute of Biology and Medicine”, Taras Shevchenko National University of Kyiv, 2, Prospekt Hlushkov, Kyiv, 03022, Ukraine
| |
Collapse
|
26
|
Bubnov R, Spivak M. Pathophysiology-Based Individualized Use of Probiotics and Prebiotics for Metabolic Syndrome: Implementing Predictive, Preventive, and Personalized Medical Approach. ADVANCES IN PREDICTIVE, PREVENTIVE AND PERSONALISED MEDICINE 2023:133-196. [DOI: 10.1007/978-3-031-19564-8_6] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
27
|
Angulo M, Ramos A, Reyes-Becerril M, Guerra K, Monreal-Escalante E, Angulo C. Probiotic Debaryomyces hansenii CBS 8339 yeast enhanced immune responses in mice. 3 Biotech 2023; 13:28. [PMID: 36590244 PMCID: PMC9797638 DOI: 10.1007/s13205-022-03442-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 12/17/2022] [Indexed: 12/29/2022] Open
Abstract
This study aimed to examine the effect of Debaryomyces hansenii CBS 8339 on innate immune responses in mice. Thirty BALB/c mice were randomly treated with phosphate buffered saline (PBS) (control) and two D. hansenii (Dh) doses: Dh 10ˆ6 CFU (colony forming units) and Dh 10ˆ8 CFU daily for 15 days. Spleen, blood, and gut samples were taken on days 7 and 15. Mouse splenocytes were isolated and challenged with Escherichia coli. Immunological assays and immune-related gene expressions were performed. Serum was obtained from blood for total IgA and IgG antibody titer determination. Gut samples were taken for yeast colonization assessment. Phagocytosis, respiratory burst activity, and nitric oxide production in mice were mainly enhanced (p < 0.05) upon 7 days of D. hansenii intake at a concentration of 10ˆ8 CFU before and after bacterial challenge. Moreover, oral D. hansenii in mice upregulated (p < 0.05) gene expression of pro-inflammatory cytokines (INF-γ, IL-6 and IL-1β) before or after E. coli challenge on day 7 but downregulated (p < 0.05) on day 15. Furthermore, total serum IgG and IgA titers were higher (p < 0.05) in Dh 10ˆ8 CFU at days 7 and 15, and only at day 7, respectively, than that in the other dose and control groups. Finally, D. hansenii was detected in the gut of mice that received the treatments, suggesting that yeast survived gastrointestinal transit. Altogether, a short period (7 days) of D. hansenii CBS 8339 oral delivery improved immune innate response on mice.
Collapse
Affiliation(s)
- Miriam Angulo
- Immunology and Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, C.P. 23096 La Paz, BCS Mexico
| | - Abel Ramos
- Immunology and Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, C.P. 23096 La Paz, BCS Mexico
| | - Martha Reyes-Becerril
- Immunology and Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, C.P. 23096 La Paz, BCS Mexico
| | - Kevyn Guerra
- Immunology and Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, C.P. 23096 La Paz, BCS Mexico
| | - Elizabeth Monreal-Escalante
- Immunology and Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, C.P. 23096 La Paz, BCS Mexico
| | - Carlos Angulo
- Immunology and Vaccinology Group, Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, C.P. 23096 La Paz, BCS Mexico
| |
Collapse
|
28
|
Bozzi Cionci N, Reggio M, Baffoni L, Di Gioia D. Probiotic Administration for the Prevention and Treatment of Gastrointestinal, Metabolic and Neurological Disorders. ADVANCES IN PREDICTIVE, PREVENTIVE AND PERSONALISED MEDICINE 2023:219-250. [DOI: 10.1007/978-3-031-19564-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
29
|
Singh S, Singh M, Gaur S. Probiotics as multifaceted oral vaccines against colon cancer: A review. Front Immunol 2022; 13:1002674. [PMID: 36263037 PMCID: PMC9573965 DOI: 10.3389/fimmu.2022.1002674] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/01/2022] [Indexed: 11/13/2022] Open
Abstract
Probiotics are known as the live microorganisms that, upon adequate administration, elicit a health beneficial response inside the host. The probiotics are known as immunomodulators and exhibit anti-tumor properties. Advanced research has explored the potential use of probiotics as the oral vaccines without the latent risks of pathogenicity. Probiotic-based oral vaccines are known to induce mucosal immunity that prevents the host from several enteric infections. Probiotic bacteria have the ability to produce metabolites in the form of anti-inflammatory cytokines, which play an important role in the prevention of carcinogenesis and in the activation of the phagocytes that eliminate the preliminary stage cancer cells. This review discusses the advantages and disadvantages of using the oral probiotic vaccines as well as the mechanism of action of probiotics in colon cancer therapy. This review also employs the use of “PROBIO” database for selecting certain probiotics with immunomodulatory properties. Furthermore, the use of several probiotic bacteria as anti-colon cancer adjuvants has also been discussed in detail. Because the current studies and trials are more focused on using the attenuated pathogens instead of using the probiotic-based vaccines, future studies must involve the advanced research in exploiting the potential of several probiotic strains as adjuvants in cancer therapies.
Collapse
Affiliation(s)
- Shubhi Singh
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
| | - Manisha Singh
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Smriti Gaur
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, India
- *Correspondence: Smriti Gaur,
| |
Collapse
|
30
|
Abstract
BACKGROUND To collect the published trials of probiotics in the treatment of diarrhea and to strictly evaluate and systematically analyze the efficacy of probiotics use for the prevention and treatment of patients with diarrhea. METHODS We searched domestic and foreign literature published between January 2016 and July 2022 to find randomized control trials that used probiotics to treat diarrhea. Only studies published in English were considered. The quality of the included literatures was assessed by using the methods provided in the Cochrane Handbook. Valid data were extracted and analyzed by meta- analysis using the Software RevMan5.2. RESULTS Total 16 trials and 1585 patients were included. The results of the meta- analysis showed that in comparison with the simple Western medicine treatment group or placebo, the added use of probiotics could improve stool frequency, stool morphology, and related irritable bowel syndrome symptoms. CONCLUSION The added use of probiotics can further improve clinical outcomes in the patients with diarrhea; however, the implementation of larger and higher quality clinical trials is necessary to verify this conclusion.
Collapse
Affiliation(s)
- Fujie Wang
- Nutritional Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ting Zhao
- Nutritional Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Weiwei Wang
- Department of Critical Care Medicine, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qianqian Dai
- Nutritional Department, Xuzhou Cancer Hospital, Xuzhou China
| | - Xianghua Ma
- Nutritional Department, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Xianghua Ma, Nutritional Department, The First Affiliated Hospital of Nanjing Medical University, No. 300, Guangzhou Road, Nanjing, Jiangsu, China (e-mail: )
| |
Collapse
|
31
|
Wielgosz-Grochowska JP, Domanski N, Drywień ME. Efficacy of an Irritable Bowel Syndrome Diet in the Treatment of Small Intestinal Bacterial Overgrowth: A Narrative Review. Nutrients 2022; 14:nu14163382. [PMID: 36014888 PMCID: PMC9412469 DOI: 10.3390/nu14163382] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/09/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
Small intestinal bacterial overgrowth (SIBO) is highly prevalent in irritable bowel syndrome (IBS). The eradication of bacterial overgrowth with antibiotics is the first-line treatment. However, focusing only on the antimicrobial effects without taking care to improve lifestyle factors, especially dietary patterns, may predispose patients to intestinal microbiota dysfunction. The objective of this study is to determine whether the current recommendations regarding nutrition in IBS are suitable for patients with SIBO. A narrative literature review was carried out using databases, including PubMed, ScienceDirect and Google Scholar. Recent studies indicate that dietary manipulation may have a role in alleviating SIBO gastrointestinal symptoms. A low FODMAP diet proposed for IBS may promote a negative shift in the gut microbiota and deepen the existing state of dysbiosis in SIBO patients. Supplementation with soluble fiber can lessen the symptoms in IBS and SIBO. Targeted probiotic therapy may also increase the effectiveness of antibiotic treatment and regulate bowel movements. Therefore, optimal dietary patterns play a key role in the treatment of SIBO. Based on currently available literature, the potential efficacy of the IBS diet in SIBO is largely hypothetical. Future research is needed to characterize a specific diet for the treatment of SIBO.
Collapse
Affiliation(s)
- Justyna Paulina Wielgosz-Grochowska
- Department of Human Nutrition, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
- Correspondence:
| | - Nicole Domanski
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Małgorzata Ewa Drywień
- Department of Human Nutrition, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
| |
Collapse
|
32
|
Ahmad Alwi NA, Lim SM, Mani V, Ramasamy K. Lactobacillus Spp.-Enhanced Memory is Strain-Dependent and Associated, in Part, with Amyloidogenic and anti-Oxidant/Oxidative Stress Interplay in Amyloid Beta Precursor Protein Transgenic Mice. J Diet Suppl 2022:1-18. [PMID: 35876040 DOI: 10.1080/19390211.2022.2103608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2022]
Abstract
This study explored mechanisms underpinning enhanced memory in amyloid precursor protein (APP) transgenic mice (male; 10-12 months; n = 6/group) supplemented with Lactobacillus plantarum LAB12 (LAB12)/Lactobacillus casei Shirota (LcS). Morris Water Maze test was performed before brains were harvested for gene expression and biochemical studies. LAB-supplemented mice exhibited reduced escape latency and distance but significant increased time spent in platform zone. This was associated with downregulated beta-site APP cleaving enzyme-1 (BACE1) mRNA and significant reduced nitric oxide in brains. LAB12 also significantly increased glutathione. The LAB-enhanced memory is strain-dependent and could be mediated, in part, through amyloidogenic pathway and anti-oxidant/oxidative stress interplay.
Collapse
Affiliation(s)
- Nor Amalina Ahmad Alwi
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Siong Meng Lim
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| | - Vasudevan Mani
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraidah, Kingdom of Saudi Arabia
| | - Kalavathy Ramasamy
- Collaborative Drug Discovery Research (CDDR) Group, Faculty of Pharmacy, Universiti Teknologi MARA (UiTM), Bandar Puncak Alam, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
33
|
Ferulic acid improves intestinal barrier function through altering gut microbiota composition in high-fat diet-induced mice. Eur J Nutr 2022; 61:3767-3783. [PMID: 35732902 DOI: 10.1007/s00394-022-02927-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 05/31/2022] [Indexed: 12/16/2022]
Abstract
PURPOSE A high-fat diet (HFD) induces gut microbiota (GM) disorders, leading to intestinal barrier dysfunction and inflammation. Ferulic acid (FA) has shown anti-obesity effects, e.g., reducing body weight and food intake. However, the mechanism linking the anti-obesity effects of FA and GM modulation remains obscure. The present study aimed to clarify the mechanism underlying the anti-obesity effects of FA and modulation of the GM. METHODS C57BL/6 J mice were fed by a low-fat diet (LFD) and HFD with or without FA at a dose of 100 mg/kg of body weight by oral gavage for 12 weeks. Using high-throughput sequencing, gas chromatography, real-time fluorescence quantitative PCR and immunohistochemical staining, the attenuation of obesity by FA were assessed via intestinal barrier integrity, inflammation, and the GM. RESULTS FA reduced weight gain, improved HFD-induced GM imbalance, significantly enhanced intestinal short-chain fatty acid (SCFA)-producing bacteria (e.g., Olsenella, Eisenbergiella, Dubosiella, Clostridiales_unclassified, and Faecalibaculum) along with SCFA accumulation and its receptors' expression, decreased endotoxin-producing bacteria or obesity-related bacterial genera, and serum endotoxin (lipopolysaccharides), and inhibited the colonic TLR4/NF-κB pathway. Thus, FA can mitigate colonic barrier dysfunction and intestinal inflammation, induce the production of SCFAs and inhibit endotoxins by modulating the GM. CONCLUSION These results indicate that enhancement of intestinal barrier by altering the GM may be an anti-obesity target of FA and that FA can be used as a functional compound with great developmental values.
Collapse
|
34
|
De Jesus LCL, Aburjaile FF, Sousa TDJ, Felice AG, Soares SDC, Alcantara LCJ, Azevedo VADC. Genomic Characterization of Lactobacillus delbrueckii Strains with Probiotics Properties. FRONTIERS IN BIOINFORMATICS 2022; 2:912795. [PMID: 36304288 PMCID: PMC9580953 DOI: 10.3389/fbinf.2022.912795] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/16/2022] [Indexed: 01/22/2023] Open
Abstract
Probiotics are health-beneficial microorganisms with mainly immunomodulatory and anti-inflammatory properties. Lactobacillus delbrueckii species is a common bacteria used in the dairy industry, and their benefits to hosting health have been reported. This study analyzed the core genome of nine strains of L. delbrueckii species with documented probiotic properties, focusing on genes related to their host health benefits. For this, a combined methodology including several software and databases (BPGA, SPAAN, BAGEL4, BioCyc, KEEG, and InterSPPI) was used to predict the most important characteristics related to L. delbrueckii strains probiose. Comparative genomics analyses revealed that L. delbrueckii probiotic strains shared essential genes related to acid and bile stress response and antimicrobial activity. Other standard features shared by these strains are surface layer proteins and extracellular proteins-encoding genes, with high adhesion profiles that interacted with human proteins of the inflammatory signaling pathways (TLR2/4-MAPK, TLR2/4-NF-κB, and NOD-like receptors). Among these, the PrtB serine protease appears to be a strong candidate responsible for the anti-inflammatory properties reported for these strains. Furthermore, genes with high proteolytic and metabolic activity able to produce beneficial metabolites, such as acetate, bioactive peptides, and B-complex vitamins were also identified. These findings suggest that these proteins can be essential in biological mechanisms related to probiotics’ beneficial effects of these strains in the host.
Collapse
Affiliation(s)
- Luís Cláudio Lima De Jesus
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Flávia Figueira Aburjaile
- Department of Preventive Veterinary Medicine, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Thiago De Jesus Sousa
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andrei Giacchetto Felice
- Department of Immunology, Microbiology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Siomar De Castro Soares
- Department of Immunology, Microbiology and Parasitology, Federal University of Triângulo Mineiro, Uberaba, Brazil
| | - Luiz Carlos Junior Alcantara
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
- Flavivirus Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
- *Correspondence: Luiz Carlos Junior Alcantara, ; Vasco Ariston De Carvalho Azevedo,
| | - Vasco Ariston De Carvalho Azevedo
- Department of Genetics, Ecology and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
- *Correspondence: Luiz Carlos Junior Alcantara, ; Vasco Ariston De Carvalho Azevedo,
| |
Collapse
|
35
|
Lin CW, Chen YT, Ho HH, Kuo YW, Lin WY, Chen JF, Lin JH, Liu CR, Lin CH, Yeh YT, Chen CW, Huang YF, Hsu CH, Hsieh PS, Yang SF. Impact of the food grade heat-killed probiotic and postbiotic oral lozenges in oral hygiene. Aging (Albany NY) 2022; 14:2221-2238. [PMID: 35236778 PMCID: PMC8954981 DOI: 10.18632/aging.203923] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/22/2022] [Indexed: 11/25/2022]
Abstract
The oral cavity plays a crucial role in food digestion and immune protection. Thus, maintaining oral health is necessary. Postbiotic and heat-killed probiotic cells have shown increased antibacterial potential with stable viability compared with live strains. However, clinical evidence regarding their effect on oral health is insufficient. Therefore, in this study, we tested postbiotic lozenges of Lactobacillus salivarius subsp. salicinius AP-32, L. paracasei ET-66, and L. plantarum LPL28 and heat-killed probiotic lozenges of L. salivarius subsp. salicinius AP-32 and L. paracasei ET-66 for their effect on oral health. In total, 75 healthy individuals were blindly and randomly divided into placebo, postbiotic lozenge, and heat-killed probiotic lozenge groups and were administered the respective lozenge type for 4 weeks. Postbiotic and heat-killed probiotic lozenge groups demonstrated antibacterial activities with a considerable increase in L. salivarius in their oral cavity. Furthermore, their salivary immunoglobulin A, Lactobacillus, and Bifidobacterium increased. Subjective questionnaires completed by the participants indicated that participants in both the experimental groups developed better oral health and intestinal conditions than those in the placebo group. Overall, our study revealed that a food additive in the form of an oral postbiotic or heat-killed probiotic lozenge may effectively enhance oral immunity, inhibit the growth of oral pathogens, and increase the numbers of beneficial oral microbiota.
Collapse
Affiliation(s)
- Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan.,Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yi-Tzu Chen
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan.,Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Hsieh-Hsun Ho
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Yi-Wei Kuo
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Wen-Yang Lin
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Jui-Fen Chen
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Jia-Hung Lin
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Cheng-Ruei Liu
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Chi-Huei Lin
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Yao-Tsung Yeh
- Aging and Disease Prevention Research Center, Fooyin University, Kaohsiung, Taiwan
| | - Ching-Wei Chen
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Yu-Fen Huang
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Chen-Hung Hsu
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Pei-Shan Hsieh
- Research and Development Department, Bioflag Biotech Co., Ltd., Tainan, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
36
|
Mirza Alizadeh A, Hosseini H, Mollakhalili Meybodi N, Hashempour-Baltork F, Alizadeh-Sani M, Tajdar-Oranj B, Pirhadi M, Mousavi Khaneghah A. Mitigation of potentially toxic elements in food products by probiotic bacteria: A comprehensive review. Food Res Int 2022; 152:110324. [PMID: 35181105 DOI: 10.1016/j.foodres.2021.110324] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 03/08/2021] [Accepted: 03/11/2021] [Indexed: 12/17/2022]
Abstract
Potentially toxic elements (PTEs) as non-degradable elements (especially carcinogenic types for humans such as lead (Pb), cadmium (Cd), mercury (Hg), and arsenic (As)) are widely distributed in the environment. They are one of the most concerned pollutants that can be absorbed and accumulated in the human body, primarily via contaminated water and foods. Acute or chronic poisoning of humans to PTEs can pose some serious risks for human health even at low concentrations. In this context, some methods are introduced to eliminate or reduce their concentration. While the biological treatment by bacterial strains, particularly probiotic bacteria, is considered as an effective method for reducing or eliminating of them. The consumption of probiotics as nonpathogenic microorganisms at regular and adequate dose offer some beneficial health impacts, it can also be applied to remove PTEs in both alive and non-alive states. This review aimed to provide an overview regarding the efficacy of different types of probiotic bacteria for PTEs removal from various environments such as food, water, in vitro, and in vivo conditions.
Collapse
Affiliation(s)
- Adel Mirza Alizadeh
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hedayat Hosseini
- Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Food Safety Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Neda Mollakhalili Meybodi
- Department of Food Sciences and Technology, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fataneh Hashempour-Baltork
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahmood Alizadeh-Sani
- Division of Food Safety and Hygiene, Environmental Health Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrouz Tajdar-Oranj
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohadeseh Pirhadi
- Division of Food Safety and Hygiene, Environmental Health Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Mousavi Khaneghah
- Department of Food Science, Faculty of Food Engineering, State University of Campinas (UNICAMP), 13083-862 Campinas, São Paulo, Brazil
| |
Collapse
|
37
|
Evangelista AG, Danielski GM, Corrêa JAF, Cavalari CMDA, Souza IR, Luciano FB, Macedo REFD. Carnobacterium as a bioprotective and potential probiotic culture to improve food quality, food safety, and human health - a scoping review. Crit Rev Food Sci Nutr 2022; 63:6946-6959. [PMID: 35156482 DOI: 10.1080/10408398.2022.2038079] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It is well-known that some bacteria can promote human and animal health. Bacteria of the genus Carnobacterium, while underexplored, have demonstrated significant probiotic and bioprotective potential. In this review, the recent scientific advances in this area are discussed. There are several requirements for a strain to be considered a probiotic or bioprotective agent, including the absence of antimicrobial resistance and the ability to colonize the gastrointestinal tract. Several researchers have reported such features in Carnobacterium bacteria, especially with regard to the production of antimicrobial substances. Research into animal production has advanced, especially in the aquaculture field, wherein inhibitory activity has been demonstrated against several important pathogens (for example Vibrio), and improvement in zootechnical indexes is evident. With respect to human health-related applications, research is still in the early stages. However, excellent in vitro results against pathogens, such as Candida albicans and Pseudomonas aeruginosa, have been reported. Carnobacterium bacteria have been assessed for a variety of applications in food, including direct application to the matrix and application to smart packaging, with proven effectiveness against Listeria monocytogenes. However, there is a lack of in vivo studies on Carnobacterium applications, which hinders its applications in various industries despite its high potential.
Collapse
Affiliation(s)
| | - Gabriela Maia Danielski
- Graduate Program in Animal Science, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil
- Undergraduate Program in Agronomy, Universidade Federal do Paraná, Curitiba, Paraná, Brazil
| | | | | | - Isabelle Ramos Souza
- Undergraduate Program in Veterinary Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, Brazil
| | | | | |
Collapse
|
38
|
Philips CA, Augustine P, Ganesan K, Ranade S, Chopra V, Patil K, Shende S, Ahamed R, Kumbar S, Rajesh S, George T, Mohanan M, Mohan N, Phadke N, Rani M, Narayanan A, Jagan SM. The role of gut microbiota in clinical complications, disease severity, and treatment response in severe alcoholic hepatitis. Indian J Gastroenterol 2022; 41:37-51. [PMID: 34989986 DOI: 10.1007/s12664-021-01157-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 02/01/2021] [Indexed: 02/04/2023]
Abstract
BACKGROUND Dysbiotic gut bacteria engage in the development and progression of severe alcoholic hepatitis (SAH). We aimed to characterize bacterial communities associated with clinical events (CE), identify significant bacteria linked to CE, and define bacterial relationships associated with specific CE and outcomes at baseline and after treatment in SAH. METHODS We performed 16-s rRNA sequencing on stool samples (n=38) collected at admission and the last follow-up within 90 days in SAH patients (n=26; 12 corticosteroids; 14 granulocyte colony-stimulating factor, [G-CSF]). Validated pipelines were used to plot bacterial communities, profile functional metabolism, and identify significant taxa and functional metabolites. Conet/NetworkX® was utilized to identify significant non-random patterns of bacterial co-presence and mutual exclusion for clinical events. RESULTS All the patients were males with median discriminant function (DF) 64, Child-Turcotte-Pugh (CTP) 12, and model for end-stage liver disease (MELD) score 25.5. At admission, 27%, 42%, and 58% had acute kidney injury (AKI), hepatic encephalopathy (HE), and infections respectively; 38.5% died at end of follow-up. Specific bacterial families were associated with HE, sepsis, disease severity, and death. Lachnobacterium and Catenibacterium were associated with HE, and Pediococcus with death after steroid treatment. Change from Enterococcus (promotes AH) to Barnesiella (inhibits E. faecium) was significant after G-CSF. Phenylpropanoid-biosynthesis (innate-immunity) and glycerophospholipid-metabolism (cellular-integrity) pathways in those without infections and the death, respectively, were upregulated. Mutual interactions between Enterococcus cecorum, Acinetobacter schindleri, and Mitsuokella correlated with admission AKI. CONCLUSIONS Specific gut microbiota, their interactions, and metabolites are associated with complications of SAH and treatment outcomes. Microbiota-based precision medicine as adjuvant treatment may be a new therapeutic area.
Collapse
Affiliation(s)
- Cyriac Abby Philips
- The Liver Unit and Monarch Liver Lab, Cochin Gastroenterology Group, Ernakulam Medical Center, Kochi, 682 028, India.
- Philip Augustine Associates (P) Ltd, Ernakulam Medical Center, Room no: 3. Ground Floor, Kochi, 682 028, India.
| | - Philip Augustine
- Gastroenterology and Advanced G.I. Endoscopy, Cochin Gastroenterology Group, Ernakulam Medical Center, Kochi, 682 028, India
| | - Karthik Ganesan
- Biomedical Software and Instrumentation, Department of Bioinformatics, Helicalbio, Ann Arbor, MI, USA
| | - Shatakshi Ranade
- Molecular, Cellular and Developmental Biology, Genepath-Dx, Pune, 411 004, India
| | - Varun Chopra
- Molecular, Cellular and Developmental Biology, Genepath-Dx, Pune, 411 004, India
| | - Kunal Patil
- Molecular, Cellular and Developmental Biology, Genepath-Dx, Pune, 411 004, India
| | - Sonie Shende
- Molecular, Cellular and Developmental Biology, Genepath-Dx, Pune, 411 004, India
| | - Rizwan Ahamed
- Gastroenterology and Advanced G.I. Endoscopy, Cochin Gastroenterology Group, Ernakulam Medical Center, Kochi, 682 028, India
| | - Sandeep Kumbar
- Gastroenterology and Advanced G.I. Endoscopy, Cochin Gastroenterology Group, Ernakulam Medical Center, Kochi, 682 028, India
| | - Sasidharan Rajesh
- Interventional Radiology, The Liver Unit and Gastroenterology, Cochin Gastroenterology Group, Ernakulam Medical Center, Kochi, 682 028, India
| | - Tom George
- Interventional Radiology, The Liver Unit and Gastroenterology, Cochin Gastroenterology Group, Ernakulam Medical Center, Kochi, 682 028, India
| | - Meera Mohanan
- Anaesthesia and Critical Care, Cochin Gastroenterology Group, Ernakulam Medical Center, Kochi, 682 028, India
| | - Narain Mohan
- The Liver Unit and Monarch Liver Lab, Cochin Gastroenterology Group, Ernakulam Medical Center, Kochi, 682 028, India
| | - Nikhil Phadke
- Molecular, Cellular and Developmental Biology, Genepath-Dx, Pune, 411 004, India
| | - Mridula Rani
- Molecular, Cellular and Developmental Biology, Genepath-Dx, Pune, 411 004, India
| | - Arjun Narayanan
- National Urban Health Mission, Ernakulam District Hospital, Kochi, 682 011, India
| | - Suchetha M Jagan
- National Urban Health Mission, Ernakulam District Hospital, Kochi, 682 011, India
| |
Collapse
|
39
|
Song X, Wang L, Liu Y, Zhang X, Weng P, Liu L, Zhang R, Wu Z. The gut microbiota–brain axis: Role of the gut microbial metabolites of dietary food in obesity. Food Res Int 2022; 153:110971. [DOI: 10.1016/j.foodres.2022.110971] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 01/27/2022] [Accepted: 01/30/2022] [Indexed: 12/13/2022]
|
40
|
Aganetti MA, Cruz CS, Galvão I, Engels DF, Ricci MF, Vieira AT. The Gut Microbiota and Immunopathophysiology. COMPREHENSIVE PHARMACOLOGY 2022:492-514. [DOI: 10.1016/b978-0-12-820472-6.00128-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
41
|
Qi C, Zhou J, Tu H, Tu R, Chang H, Chen J, Li D, Sun J, Yu R. Lactation-dependent vertical transmission of natural probiotics from the mother to the infant gut through breast milk. Food Funct 2021; 13:304-315. [PMID: 34889924 DOI: 10.1039/d1fo03131g] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The transmission of certain bacteria from the mother's gut to the infant's gut via breast milk (BM) is critical for the offspring's immune system development. Dysbiosis of the BM microbiota can be caused by a variety of reasons, which can be influenced by probiotics delivered via the enteromammary route. The goal of this study was to investigate the bacteria that can be transmitted from the mother to the infant's intestine during various lactation periods in 19 mother-child dyads. Bacterial transmission is most common during the colostrum phase when bacteria with certain amplicon sequence variants (ASVs) enter the newborn intestine and inhabit it permanently. We have established that anaerobic gut-associated bacteria, such as Faecalibacterium, Blautia and Lachnoclostridium, transfer from the mother to the infant's gut with lactation dependence using the idea of weighted transfer ratios. Streptococcus salivarius, Bifidobacterium longum, and Lactobacillus gasseri are transferred from the maternal gut to the BM, as well as from the BM to the newborn gut, depending on different ASVs. These findings suggest that isolation of key microorganisms from breast milk could be utilized to modify the microbiota of BM or newborns by giving the mother a probiotic or adding it to artificial milk to promote neonatal health.
Collapse
Affiliation(s)
- Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| | - Jingbo Zhou
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| | - Huayu Tu
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| | - Rundan Tu
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| | - Hong Chang
- Department of Pediatric Cardiology, Nephrology and Rheumatism, The Affiliated Hospital of Qingdao University Medical College, Qingdao 266003, PR China
| | - Jie Chen
- Department of Obstetrics, Affiliated Hospital of Qingdao University, Qingdao 266000, PR China
| | - Duo Li
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| | - Renqiang Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, PR China. E-mail:.
| |
Collapse
|
42
|
L M L, L P B, S G G, L O S, O M D, R V B, L M S, M Ya S. Assessment of the Safety of Lactobacillus casei IMV B-7280 Probiotic Strain on a Mouse Model. Probiotics Antimicrob Proteins 2021; 13:1644-1657. [PMID: 33876388 PMCID: PMC8055307 DOI: 10.1007/s12602-021-09789-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2021] [Indexed: 01/19/2023]
Abstract
Probiotics, in particular Lactobacillus (lactic acid bacteria, LAB) strains, are widely used in clinical practice. Despite that these probiotics have GRAS (generally regarded as safe) and qualified presumption of safety (QPS) statuses, the safety of particular strains still needs to be thoroughly studied. The aim of the study was to evaluate the safety of Lact. casei IMV B-7280 strain by investigating toxicity and the effects on gut microbiota in experimental animal model. Male BALB/c mice (7-8 weeks, weight 20-24 g) were treated with amounts of Lact. casei IMV B-7280 strain: 5 × 106, 5 × 108, or 5 × 109 CFU/animal once per day during 7 days, or in the amount of 1 × 1010 CFU/animal once per day during 3 days (most of the proposed probiotic doses for humans-from 108 to 109 CFU) and monitored during 14 days. Blood tests and serum biochemistry were conducted; the cecal content from mice of the experimental and control groups were freshly collected and analyzed. At the end of the experiments (15th day), the presence of LAB in the heart, liver, kidney, and mesenteric lymph nodes and peripheral blood was determined; histology of the brain, liver, heart, fragments of the small and large intestine, and mesenteric lymph nodes was conducted. Survival rate of BALB/c mice treated with Lact. casei IMV B-7280 strain in different concentrations in toxicity experiments during 14 days was 100%. We observed no signs of toxicity as changes in gait, lethargy, sleep, somatomotor activity as well as changes in fur, eyes, skin and mucous membranes, tremors, behavior pattern, convulsions, salivation, diarrhea, and local injuries in mice from all experimental groups. After administration of probiotic strain, the number of opportunistic bacteria in cecal contents, such as Staphylococcus spp., Candida spp., Pseudomonas spp., and total aerobic and optionally anaerobic bacteria decreased compared to controls; the population of beneficial bacteria such as lactobacilli increased in cecal contents of these mice. LAB were not detected in the peripheral blood, heart, liver, kidneys, and mesenteric lymph nodes after administration of this strain to intact mice. Lact. casei IMV B-7280 strain is safe at dose up to 1010 CFU/animal during 3- and 7-day oral administration to mice and has a positive effect on the gut microbiota composition; it could be potentially considered as safe probiotic for humans.
Collapse
Affiliation(s)
- Lazarenko L M
- Zabolotny Institute of microbiology and virology, National Academy of Sciences of Ukraine, 154, Akad. Zabolotny str, Kyiv, 03143, Ukraine
| | - Babenko L P
- Zabolotny Institute of microbiology and virology, National Academy of Sciences of Ukraine, 154, Akad. Zabolotny str, Kyiv, 03143, Ukraine
| | - Gichka S G
- Bogomolets National Medical University, 13, T. Shevchenko blvd, Kyiv, 01601, Ukraine
| | - Sakhno L O
- RE Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, National Academy of Sciences of Ukraine, 45, Vasylkivska str, Kyiv, 03022, Ukraine
| | - Demchenko O M
- Zabolotny Institute of microbiology and virology, National Academy of Sciences of Ukraine, 154, Akad. Zabolotny str, Kyiv, 03143, Ukraine
| | - Bubnov R V
- Zabolotny Institute of microbiology and virology, National Academy of Sciences of Ukraine, 154, Akad. Zabolotny str, Kyiv, 03143, Ukraine.
| | - Sichel L M
- Pure Research Products, LLC, 6107 Chelsea Manor Court, Boulder, Colorado, 80301, USA
| | - Spivak M Ya
- Zabolotny Institute of microbiology and virology, National Academy of Sciences of Ukraine, 154, Akad. Zabolotny str, Kyiv, 03143, Ukraine
| |
Collapse
|
43
|
Singh R, Zogg H, Ro S. Role of microRNAs in Disorders of Gut-Brain Interactions: Clinical Insights and Therapeutic Alternatives. J Pers Med 2021; 11:1021. [PMID: 34683162 PMCID: PMC8541612 DOI: 10.3390/jpm11101021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
Disorders of gut-brain interactions (DGBIs) are heterogeneous in nature and intertwine with diverse pathophysiological mechanisms. Regular functioning of the gut requires complex coordinated interplay between a variety of gastrointestinal (GI) cell types and their functions are regulated by multiple mechanisms at the transcriptional, post-transcriptional, translational, and post-translational levels. MicroRNAs (miRNAs) are small non-coding RNA molecules that post-transcriptionally regulate gene expression by binding to specific mRNA targets to repress their translation and/or promote the target mRNA degradation. Dysregulation of miRNAs might impair gut physiological functions leading to DGBIs and gut motility disorders. Studies have shown miRNAs regulate gut functions such as visceral sensation, gut immune response, GI barrier function, enteric neuronal development, and GI motility. These biological processes are highly relevant to the gut where neuroimmune interactions are key contributors in controlling gut homeostasis and functional defects lead to DGBIs. Although extensive research has explored the pathophysiology of DGBIs, further research is warranted to bolster the molecular mechanisms behind these disorders. The therapeutic targeting of miRNAs represents an attractive approach for the treatment of DGBIs because they offer new insights into disease mechanisms and have great potential to be used in the clinic as diagnostic markers and therapeutic targets. Here, we review recent advances regarding the regulation of miRNAs in GI pacemaking cells, immune cells, and enteric neurons modulating pathophysiological mechanisms of DGBIs. This review aims to assess the impacts of miRNAs on the pathophysiological mechanisms of DGBIs, including GI dysmotility, impaired intestinal barrier function, gut immune dysfunction, and visceral hypersensitivity. We also summarize the therapeutic alternatives for gut microbial dysbiosis in DGBIs, highlighting the clinical insights and areas for further exploration. We further discuss the challenges in miRNA therapeutics and promising emerging approaches.
Collapse
Affiliation(s)
| | | | - Seungil Ro
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, 1664 North Virginia Street, Reno, NV 89557, USA; (R.S.); (H.Z.)
| |
Collapse
|
44
|
Golubnitschaja O, Liskova A, Koklesova L, Samec M, Biringer K, Büsselberg D, Podbielska H, Kunin AA, Evsevyeva ME, Shapira N, Paul F, Erb C, Dietrich DE, Felbel D, Karabatsiakis A, Bubnov R, Polivka J, Polivka J, Birkenbihl C, Fröhlich H, Hofmann-Apitius M, Kubatka P. Caution, "normal" BMI: health risks associated with potentially masked individual underweight-EPMA Position Paper 2021. EPMA J 2021; 12:243-264. [PMID: 34422142 PMCID: PMC8368050 DOI: 10.1007/s13167-021-00251-4] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 07/30/2021] [Indexed: 02/06/2023]
Abstract
An increasing interest in a healthy lifestyle raises questions about optimal body weight. Evidently, it should be clearly discriminated between the standardised "normal" body weight and individually optimal weight. To this end, the basic principle of personalised medicine "one size does not fit all" has to be applied. Contextually, "normal" but e.g. borderline body mass index might be optimal for one person but apparently suboptimal for another one strongly depending on the individual genetic predisposition, geographic origin, cultural and nutritional habits and relevant lifestyle parameters-all included into comprehensive individual patient profile. Even if only slightly deviant, both overweight and underweight are acknowledged risk factors for a shifted metabolism which, if being not optimised, may strongly contribute to the development and progression of severe pathologies. Development of innovative screening programmes is essential to promote population health by application of health risks assessment, individualised patient profiling and multi-parametric analysis, further used for cost-effective targeted prevention and treatments tailored to the person. The following healthcare areas are considered to be potentially strongly benefiting from the above proposed measures: suboptimal health conditions, sports medicine, stress overload and associated complications, planned pregnancies, periodontal health and dentistry, sleep medicine, eye health and disorders, inflammatory disorders, healing and pain management, metabolic disorders, cardiovascular disease, cancers, psychiatric and neurologic disorders, stroke of known and unknown aetiology, improved individual and population outcomes under pandemic conditions such as COVID-19. In a long-term way, a significantly improved healthcare economy is one of benefits of the proposed paradigm shift from reactive to Predictive, Preventive and Personalised Medicine (PPPM/3PM). A tight collaboration between all stakeholders including scientific community, healthcare givers, patient organisations, policy-makers and educators is essential for the smooth implementation of 3PM concepts in daily practice.
Collapse
Affiliation(s)
- Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Alena Liskova
- Clinic of Obstetrics and Gynaecology, Jessenius Faculty of Medicine, Comenius University, in Bratislava, 03601 Martin, Slovakia
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynaecology, Jessenius Faculty of Medicine, Comenius University, in Bratislava, 03601 Martin, Slovakia
| | - Marek Samec
- Clinic of Obstetrics and Gynaecology, Jessenius Faculty of Medicine, Comenius University, in Bratislava, 03601 Martin, Slovakia
| | - Kamil Biringer
- Clinic of Obstetrics and Gynaecology, Jessenius Faculty of Medicine, Comenius University, in Bratislava, 03601 Martin, Slovakia
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, 24144 Doha, Qatar
| | - Halina Podbielska
- Department of Biomedical Engineering, Faculty of Fundamental Problems of Technology, Wrocław University of Science and Technology, 50-370 Wrocław, Poland
| | - Anatolij A. Kunin
- Departments of Maxillofacial Surgery and Hospital Dentistry, Voronezh N.N. Burdenko State Medical University, Voronezh, Russian Federation
| | | | - Niva Shapira
- Nutrition Department, Ashkelon Academic College, Ashkelon, Tel Aviv, Israel
| | - Friedemann Paul
- NeuroCure Clinical Research Centre, Experimental and Clinical Research Centre, Max Delbrueck Centre for Molecular Medicine and Charité Universitaetsmedizin Berlin, Berlin, Germany
| | - Carl Erb
- Private Institute of Applied Ophthalmology, Berlin, Germany
| | - Detlef E. Dietrich
- European Depression Association, Brussels, Belgium
- AMEOS Clinical Centre for Psychiatry and Psychotherapy, 31135 Hildesheim, Germany
| | - Dieter Felbel
- Fachklinik Kinder und Jugendliche Psychiatrie, AMEOS Klinikum Hildesheim, Akademisches Lehrkrankenhaus für Pflege der FOM Hochschule Essen, Hildesheim, Germany
| | - Alexander Karabatsiakis
- Institute of Psychology, Department of Clinical Psychology II, University of Innsbruck, Innsbruck, Austria
| | - Rostyslav Bubnov
- Ultrasound Department, Clinical Hospital “Pheophania”, Kyiv, Ukraine
- Zabolotny Institute of Microbiology and Virology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Jiri Polivka
- Department of Neurology, Faculty of Medicine in Pilsen, Charles University and University Hospital Pilsen, Pilsen, Czech Republic
| | - Jiri Polivka
- Department of Histology and Embryology, Faculty of Medicine in Pilsen, Charles University, Staré Město, Czech Republic
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Staré Město, Czech Republic
| | - Colin Birkenbihl
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53757 Sankt Augustin, Germany
- Bonn-Aachen International Centre for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
| | - Holger Fröhlich
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53757 Sankt Augustin, Germany
- Bonn-Aachen International Centre for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
- UCB Biosciences GmbH, Alfred-Nobel Str. 10, 40789 Monheim am Rhein, Germany
| | - Martin Hofmann-Apitius
- Department of Bioinformatics, Fraunhofer Institute for Algorithms and Scientific Computing (SCAI), Schloss Birlinghoven, 53757 Sankt Augustin, Germany
- Bonn-Aachen International Centre for IT, Rheinische Friedrich-Wilhelms-Universität Bonn, 53115 Bonn, Germany
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
45
|
Kim SJ, Choi SI, Jang M, Jeong YA, Kang CH, Kim GH. Combination of Limosilactobacillus fermentum MG4231 and MG4244 attenuates lipid accumulation in high-fat diet-fed obese mice. Benef Microbes 2021; 12:479-491. [PMID: 34348593 DOI: 10.3920/bm2020.0205] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We investigated the anti-obesity effect and the underlying mechanisms of action of human-derived Limosilactobacillus fermentum MG4231, MG4244, and their combination, in high-fat diet-induced obese mice. Administration of the Limosilactobacillus strains decreased body weight gain, liver and adipose tissue weight, and glucose tolerance. Serum levels of total cholesterol, low-density lipoprotein-cholesterol, and leptin were reduced, while adiponectin increased. The administration of Limosilactobacillus strains improved the histopathological features of liver tissue, such as hepatic atrophy and inflammatory penetration, and significantly reduced the content of triglyceride in the liver. Limosilactobacillus administration discovered a significant reduction in the size of the adipocytes in the epididymal tissue. Limosilactobacillus treatment significantly reduced the expression of important regulators in lipid metabolism, including peroxisome proliferator-activated receptor γ, CCAAT/enhancer-binding protein α, fatty acid synthase (FAS), adipocyte-protein 2, and lipoprotein lipase in the epididymal tissue. Also, Limosilactobacillus lowered sterol regulatory element-binding protein 1-c and FAS in the liver tissue. Such changes in the expression of these regulators in both liver and epididymis tissue were caused by Limosilactobacillus upregulating phosphorylation of AMP-activated protein kinase and acetyl-CoA carboxylase. Therefore, we suggest that the use of the combination of L. fermentum MG4231 and MG4244, as probiotics could effectively inhibit adipogenesis and lipogenesis from preventing obesity.
Collapse
Affiliation(s)
- S J Kim
- Department of Health Functional Materials, Duksung Women's University, 144 gil, Dobong-gu, Seoul, 01369, Republic of Korea
| | - S-I Choi
- Department of Health Functional Materials, Duksung Women's University, 144 gil, Dobong-gu, Seoul, 01369, Republic of Korea
| | - M Jang
- Department of Food and Life Science, Inje University, Gimhae, Republic of Korea
| | - Y-A Jeong
- R&D Center, MEDIOGEN Co., Ltd., Seoul, Republic of Korea
| | - C-H Kang
- R&D Center, MEDIOGEN Co., Ltd., Seoul, Republic of Korea
| | - G-H Kim
- Department of Food and Nutrition, Duksung Women's University, 33, Samyang-ro 144-gil, Dobong-gu, Seoul, 01369, Republic of Korea
| |
Collapse
|
46
|
Berezhnaya Y, Bikaeva I, Gachkovskaia A, Demidenko A, Klimenko N, Tyakht A, Volokh O, Alexeev D. Temporal dynamics of probiotic Lacticaseibacillus casei and rhamnosus abundance in a fermented dairy product evaluated using a combination of cultivation-dependent and -independent methods. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.111750] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
47
|
Effects of the Use of a Combination of Two Bacillus Species on Performance, Egg Quality, Small Intestinal Mucosal Morphology, and Cecal Microbiota Profile in Aging Laying Hens. Probiotics Antimicrob Proteins 2021; 12:204-213. [PMID: 30810908 DOI: 10.1007/s12602-019-09532-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sixty-week-old Hy-Line brown laying hens were randomly divided into five groups and fed different diets over a period of 84 days. Experimental treatments included a basal diet (control); the basal diet supplemented with 1.0 × 106B. licheniformis yb-214245; the basal diet supplemented with 1.0 × 106B. subtilis yb-114246; a combination of both strains in a 2:1 ratio (6.6 × 105:3.3 × 105B. licheniformis yb-214245:B. subtilis yb-114246); and the latter, added with 5 mg/kg flavomycin. Basal diet supplementation with the combined Bacillus species improved egg-laying performance in aging hens significantly (P < 0.05). Eggshell strength improved significantly with this treatment, compared to the control or the antibiotic-supplemented groups (P < 0.05). The levels of total cholesterol, triglycerides, and very low-density lipoprotein cholesterol in egg yolk declined significantly more in the Bacillus-treated group than in the control or the antibiotic-supplemented groups (P < 0.01). Small intestinal morphology was better in the hens treated with the Bacillus combination than in the hens in the control group (P < 0.05). The total number of aerobic bacteria (Bacillus, Lactobacillus, and Bifidobacterium) in the cecum was significantly higher in all the Bacillus-supplemented hens than either in the control or the antibiotic-supplemented hens (P < 0.01); additionally, the number of E. coli and Salmonella was significantly lower than in the control group (P < 0.01). In conclusion, diet supplementation with the combination of Bacillus species used here for aging laying hens improved their growth performance, cecal bacterial composition, egg quality, and small intestine morphology.
Collapse
|
48
|
Kudela E, Liskova A, Samec M, Koklesova L, Holubekova V, Rokos T, Kozubik E, Pribulova T, Zhai K, Busselberg D, Kubatka P, Biringer K. The interplay between the vaginal microbiome and innate immunity in the focus of predictive, preventive, and personalized medical approach to combat HPV-induced cervical cancer. EPMA J 2021; 12:199-220. [PMID: 34194585 PMCID: PMC8192654 DOI: 10.1007/s13167-021-00244-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/04/2021] [Indexed: 12/20/2022]
Abstract
HPVs representing the most common sexually transmitted disease are a group of carcinogenic viruses with different oncogenic potential. The immune system and the vaginal microbiome represent the modifiable and important risk factors in HPV-induced carcinogenesis. HPV infection significantly increases vaginal microbiome diversity, leading to gradual increases in the abundance of anaerobic bacteria and consequently the severity of cervical dysplasia. Delineation of the exact composition of the vaginal microbiome and immune environment before HPV acquisition, during persistent/progressive infections and after clearance, provides insights into the complex mechanisms of cervical carcinogenesis. It gives hints regarding the prediction of malignant potential. Relative high HPV prevalence in the general population is a challenge for modern and personalized diagnostics and therapeutic guidelines. Identifying the dominant microbial biomarkers of high-grade and low-grade dysplasia could help us to triage the patients with marked chances of lesion regression or progression. Any unnecessary surgical treatment of cervical dysplasia could negatively affect obstetrical outcomes and sexual life. Therefore, understanding the effect and role of microbiome-based therapies is a breaking point in the conservative management of HPV-associated precanceroses. The detailed evaluation of HPV capabilities to evade immune mechanisms from various biofluids (vaginal swabs, cervicovaginal lavage/secretions, or blood) could promote the identification of new immunological targets for novel individualized diagnostics and therapy. Qualitative and quantitative assessment of local immune and microbial environment and associated risk factors constitutes the critical background for preventive, predictive, and personalized medicine that is essential for improving state-of-the-art medical care in patients with cervical precanceroses and cervical cancer. The review article focuses on the influence and potential diagnostic and therapeutic applications of the local innate immune system and the microbial markers in HPV-related cancers in the context of 3P medicine.
Collapse
Affiliation(s)
- Erik Kudela
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 036 01 Martin, Slovakia
| | - Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 036 01 Martin, Slovakia
| | - Marek Samec
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 036 01 Martin, Slovakia
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 036 01 Martin, Slovakia
| | - Veronika Holubekova
- Jessenius Faculty of Medicine, Biomedical Centre Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Tomas Rokos
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 036 01 Martin, Slovakia
| | - Erik Kozubik
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 036 01 Martin, Slovakia
| | - Terezia Pribulova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 036 01 Martin, Slovakia
| | - Kevin Zhai
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, 24144 Doha, Qatar
| | - Dietrich Busselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, 24144 Doha, Qatar
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 036 01 Martin, Slovakia
- European Association for Predictive, Preventive and Personalised Medicine, EPMA, 1160 Brussels, Belgium
| | - Kamil Biringer
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Kollarova 2, 036 01 Martin, Slovakia
| |
Collapse
|
49
|
de Oliveira Y, Cavalcante RGS, Cavalcanti Neto MP, Magnani M, Braga VDA, de Souza EL, de Brito Alves JL. Oral administration of Lactobacillus fermentum post-weaning improves the lipid profile and autonomic dysfunction in rat offspring exposed to maternal dyslipidemia. Food Funct 2021; 11:5581-5594. [PMID: 32524104 DOI: 10.1039/d0fo00514b] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Maternal dyslipidemia alters the gut microbiota composition and contributes to the development of arterial hypertension (AH) in offspring. Here, we evaluated the effects of a new Lactobacillus fermentum probiotic formulation given post-weaning on cardiometabolic parameters and gut microbiota in male and female rat offspring from dams exposed to maternal dyslipidemia during pregnancy and lactation. METHODS Wistar rats (n = 14) were fed with a control diet (CTL = 7) or a dyslipidemic diet (DLP = 7) during pregnancy and lactation. After weaning, male and female offspring received a standard diet up to 90 days of life. Rats were allocated into three groups: CTL group + saline solution (n = 14); DLP group + saline solution (n = 14) and DLP group receiving a probiotic cocktail (n = 14). A vehicle or probiotic formulation containing L. fermentum 139, L. fermentum 263 and L. fermentum 296 (ratio 1 : 1 : 1, 1 × 109 CFU mL-1) was administered daily by oral gavage for 8 weeks. RESULTS The intervention with the probiotic formulation of L. fermentum in male and female offspring reduced total cholesterol (TC) and increased HDL-c, but did not affect the insulin resistance induced by maternal dyslipidemia. Additionally, the male and female rats that received the probiotic formulation of L. fermentum demonstrated improvement in fecal Lactobacillus sp. counts, blood pressure and sympathetic tone, without affecting baroreflex modulation. CONCLUSION The probiotic formulation containing L. fermentum improved the lipid profile and autonomic dysfunction in male and female offspring exposed to maternal dyslipidemia.
Collapse
Affiliation(s)
- Yohanna de Oliveira
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa, Brazil.
| | | | | | - Marciane Magnani
- Department of Food Engineering, Technology Center, Federal University of Paraíba, Joao Pessoa, Brazil
| | - Valdir de Andrade Braga
- Department of Biotechnology, Biotechnology Center, Federal University of Paraíba, João Pessoa, Brazil and Department of Physiology and Pharmacology, Karolinska Institute, Stockholm, Sweden
| | - Evandro Leite de Souza
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa, Brazil.
| | - José Luiz de Brito Alves
- Department of Nutrition, Health Sciences Center, Federal University of Paraíba, João Pessoa, Brazil.
| |
Collapse
|
50
|
The Role of Bifidobacteria in Predictive and Preventive Medicine: A Focus on Eczema and Hypercholesterolemia. Microorganisms 2021; 9:microorganisms9040836. [PMID: 33919907 PMCID: PMC8070932 DOI: 10.3390/microorganisms9040836] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/23/2021] [Accepted: 04/07/2021] [Indexed: 12/15/2022] Open
Abstract
Bifidobacteria colonize the human gastrointestinal tract early on in life, their interaction with the host starting soon after birth. The health benefits are strain specific and could be due to the produced polysaccharides. The consumption of probiotics may prevent obesity, irritable bowel syndrome, eczema or atopic dermatitis, and asthma. Non-replicative strains of Bifidobacterium longum (NCC3001 and NCC2705) promote the differentiation of normal human epidermal keratinocytes (NHEKs), inducing a high expression of differentiation markers (keratin —KRT1—, and transglutaminase —TGM1—) and pro-regeneration markers (cathepsins), including β-defensin-1, which plays an important role in modulating the cutaneous immune response. Strains belonging to the genera Bifidobacterium and Lactobacillus can increase tight-junction proteins in NHEKs and enhance barrier function. Bifidobacteria and lactobacilli may be used as prophylactic or therapeutic agents towards enteric pathogens, antibiotic-associated diarrhea, lactose intolerance, ulcerative colitis, irritable bowel syndrome, colorectal cancer, cholesterol reduction, and control of obesity and metabolic disorders. Bifidobacterium bifidum showed an in vitro capability of lowering cholesterol levels thanks to its absorption into the bacterial membrane. Several strains of the species Lactobacillus acidophilus, L. delbrueckii subsp. bulgaricus, L. casei, and L. gasseri led to a reduced amount of serum cholesterol due to their ability to assimilate cholesterol (in vitro). Lactococcus lactis KF147 and Lactobacillus plantarum Lp81 have also been shown to reduce cholesterol levels by 12%. Clarifying the specific health mechanisms of Bifidobacterium and Lactobacillus strains in preventing high-cost pathologies could be useful for delineating effective guidelines for the treatment of infants and adults.
Collapse
|