Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (26)

Search Parameters:
Keywords = gut microbiological profile

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
34 pages, 6295 KiB  
Article
ROS/Enzyme Dual-Responsive Drug Delivery System for Targeted Colorectal Cancer Therapy: Synergistic Chemotherapy, Anti-Inflammatory, and Gut Microbiota Modulation
by Xin Zhang, Ruonan Lian, Bingbing Fan, Lei Meng, Pengxia Zhang, Yu Zhang and Weitong Sun
Pharmaceutics 2025, 17(7), 940; https://doi.org/10.3390/pharmaceutics17070940 - 21 Jul 2025
Viewed by 420
Abstract
Objectives: Colorectal cancer (CRC) is a leading cause of cancer-related mortality, driven by chronic inflammation, gut microbiota dysbiosis, and complex tumor microenvironment interactions. Current therapies are limited by systemic toxicity and poor tumor accumulation. This study aimed to develop a ROS/enzyme dual-responsive oral [...] Read more.
Objectives: Colorectal cancer (CRC) is a leading cause of cancer-related mortality, driven by chronic inflammation, gut microbiota dysbiosis, and complex tumor microenvironment interactions. Current therapies are limited by systemic toxicity and poor tumor accumulation. This study aimed to develop a ROS/enzyme dual-responsive oral drug delivery system, KGM-CUR/PSM microspheres, to achieve precise drug release in CRC and enhance tumor-specific drug accumulation, which leverages high ROS levels in CRC and the β-mannanase overexpression in colorectal tissues. Methods: In this study, we synthesized a ROS-responsive prodrug polymer (PSM) by conjugating polyethylene glycol monomethyl ether (mPEG) and mesalazine (MSL) via a thioether bond. CUR was then encapsulated into PSM using thin-film hydration to form tumor microenvironment-responsive micelles (CUR/PSM). Subsequently, konjac glucomannan (KGM) was employed to fabricate KGM-CUR/PSM microspheres, enabling targeted delivery for colorectal cancer therapy. The ROS/enzyme dual-response properties were confirmed through in vitro drug release studies. Cytotoxicity, cellular uptake, and cell migration were assessed in SW480 cells. In vivo efficacy was evaluated in AOM/DSS-induced CRC mice, monitoring tumor growth, inflammatory markers (TNF-α, IL-1β, IL-6, MPO), and gut microbiota composition. Results: In vitro drug release studies demonstrated that KGM-CUR/PSM microspheres exhibited ROS/enzyme-responsive release profiles. CUR/PSM micelles demonstrated significant anti-CRC efficacy in cytotoxicity assays, cellular uptake studies, and cell migration assays. In AOM/DSS-induced CRC mice, KGM-CUR/PSM microspheres significantly improved survival and inhibited CRC tumor growth, and effectively reduced the expression of inflammatory cytokines (TNF-α, IL-1β, IL-6) and myeloperoxidase (MPO). Histopathological and microbiological analyses revealed near-normal colon architecture and microbial diversity in the KGM-CUR/PSM group, confirming the system’s ability to disrupt the “inflammation-microbiota-tumor” axis. Conclusions: The KGM-CUR/PSM microspheres demonstrated a synergistic enhancement of anti-tumor efficacy by inducing apoptosis, alleviating inflammation, and modulating the intestinal microbiota, which offers a promising stimuli-responsive drug delivery system for future clinical treatment of CRC. Full article
(This article belongs to the Section Drug Delivery and Controlled Release)
Show Figures

Figure 1

58 pages, 656 KiB  
Review
Human Digestive Physiology and Evolutionary Diet: A Metabolomic Perspective on Carnivorous and Scavenger Adaptations
by Vicente Javier Clemente-Suárez, Laura Redondo-Flórez, Ana Isabel Beltrán-Velasco, Rodrigo Yáñez-Sepúlveda, Alejandro Rubio-Zarapuz, Alexandra Martín-Rodríguez, Eduardo Navarro-Jimenez and José Francisco Tornero-Aguilera
Metabolites 2025, 15(7), 453; https://doi.org/10.3390/metabo15070453 - 4 Jul 2025
Viewed by 1671
Abstract
This review examines human digestive physiology and metabolic adaptations in the context of evolutionary dietary patterns, particularly those emphasizing carnivorous and scavenging behaviors. By integrating metabolomic data with archaeological, anatomical, and microbiological evidence, the study explores how early hominins adapted to intermittent but [...] Read more.
This review examines human digestive physiology and metabolic adaptations in the context of evolutionary dietary patterns, particularly those emphasizing carnivorous and scavenging behaviors. By integrating metabolomic data with archaeological, anatomical, and microbiological evidence, the study explores how early hominins adapted to intermittent but energy-dense animal-based diets. The analysis highlights the development of hepatic insulin resistance, enhanced fat and protein metabolism, and shifts in gut microbiota diversity as physiological signatures of meat consumption. Comparative evaluations of digestive enzyme profiles, intestinal morphology, and salivary composition underscore humans’ omnivorous flexibility and partial carnivorous specialization. Additionally, biomarkers such as ketone bodies, branched-chain amino acids, and trimethylamine-N-oxide are identified as metabolic indicators of habitual meat intake. These adaptations, though once evolutionarily advantageous, are discussed in relation to current metabolic disorders in modern nutritional contexts. Overall, this review presents a metabolomic framework for understanding the evolutionary trajectory of human digestion and its implications for health and dietary recommendations. Full article
(This article belongs to the Section Advances in Metabolomics)
27 pages, 980 KiB  
Review
The Role of the Gut Microbiota in Female Reproductive and Gynecological Health: Insights into Endometrial Signaling Pathways
by Patricia Escorcia Mora, Diana Valbuena and Antonio Diez-Juan
Life 2025, 15(5), 762; https://doi.org/10.3390/life15050762 - 9 May 2025
Cited by 5 | Viewed by 2637
Abstract
Fertility is a dynamic, multifactorial process governed by hormonal, immune, metabolic, and environmental factors. Recent evidence highlights the gut microbiota as a key systemic regulator of reproductive health, with notable impacts on endometrial function, implantation, pregnancy maintenance, and the timing of birth. This [...] Read more.
Fertility is a dynamic, multifactorial process governed by hormonal, immune, metabolic, and environmental factors. Recent evidence highlights the gut microbiota as a key systemic regulator of reproductive health, with notable impacts on endometrial function, implantation, pregnancy maintenance, and the timing of birth. This review examines the gut–endometrial axis, focusing on how gut microbial communities influence reproductive biology through molecular signaling pathways. We discuss the modulatory roles of microbial-derived metabolites—including short-chain fatty acids, bile acids, and tryptophan catabolites—in shaping immune tolerance, estrogen metabolism, and epithelial integrity at the uterine interface. Emphasis is placed on shared mechanisms such as β-glucuronidase-mediated estrogen recycling, Toll-like receptor (TLR)-driven inflammation, Th17/Treg cell imbalance, and microbial translocation, which collectively implicate dysbiosis in the etiology of gynecological disorders including endometriosis, polycystic ovary syndrome (PCOS), recurrent implantation failure (RIF), preeclampsia (PE), and preterm birth (PTB). Although most current evidence remains correlational, emerging insights from metagenomic and metabolomic profiling, along with microbiota-depletion models and Mendelian randomization studies, underscore the biological significance of gut-reproductive crosstalk. By integrating concepts from microbiology, immunology, and reproductive molecular biology, this review offers a systems-level perspective on host–microbiota interactions in female fertility. Full article
(This article belongs to the Section Reproductive and Developmental Biology)
Show Figures

Figure 1

16 pages, 2241 KiB  
Article
A Two-Phage Cocktail Modulates Gut Microbiota Composition and Metabolic Profiles in an Ex Vivo Colon Model
by Sthefhany Nohemí Rodríguez-Arellano, Jean Pierre González-Gómez, Bruno Gomez-Gil, Marisela González-Ávila, Juan Ramón Palomera-Hernández, Elisa Barrón-Cabrera, Marcela de Jesús Vergara-Jiménez and Cristobal Chaidez
Int. J. Mol. Sci. 2025, 26(6), 2805; https://doi.org/10.3390/ijms26062805 - 20 Mar 2025
Viewed by 740
Abstract
Bacteriophage therapy is a promising approach for targeting antibiotic-resistant bacteria and modulating gut microbiota in metabolic diseases such as obesity. This study evaluated the impact of a two-phage cocktail on an ex vivo colonic simulation model of gut microbiota derived from obese individuals, [...] Read more.
Bacteriophage therapy is a promising approach for targeting antibiotic-resistant bacteria and modulating gut microbiota in metabolic diseases such as obesity. This study evaluated the impact of a two-phage cocktail on an ex vivo colonic simulation model of gut microbiota derived from obese individuals, both in its normalized state and after enrichment with Enterobacter cloacae, an obesity-related bacteria. Microbiological analyses confirmed that the phage cocktail remained active throughout the colonic regions over three digestion cycles and effectively reduced enterobacterial populations in the enriched microbiota. Metabarcoding of the 16S rRNA gene revealed that phage therapy did not significantly alter the abundance of dominant genera, but selectively reduced E. cloacae across all colonic regions. Alpha diversity was significantly affected only in the enriched microbiota, while beta diversity analysis indicated significant compositional shifts during therapy, with reduced dispersion in the final treatment stage. Short-chain fatty acid profiling demonstrated region- and group-specific metabolic responses, with increased lactic and butyric acid concentrations in the ascending colon of the enriched microbiota following phage treatment. This study provides the first ex vivo evidence that a two-phage cocktail can selectively eliminate E. cloacae while preserving overall microbiota structure and functionality. These findings establish a foundation for future in vivo studies exploring the role of phage therapy in reshaping gut microbial communities and metabolic profiles, highlighting its potential as a precision tool for managing gut dysbiosis in metabolic disorders. Full article
(This article belongs to the Special Issue Molecular Research on Bacteria)
Show Figures

Figure 1

26 pages, 1303 KiB  
Review
The Urogenital System Microbiota: Is It a New Gamechanger in Urogenital Cancers?
by Gülfem Ece, Ahmet Aktaş, Ayse Caner, İmran Sağlık, Tuğba Kula Atik, Özlem Ulusan Bağcı, Fulya Bayındır Bilman, Hadiye Demirbakan, Seda Güdül Havuz, Esra Kaya, Özlem Koyuncu Özyurt, Gülay Yetkin and Orçun Zorbozan
Microorganisms 2025, 13(2), 315; https://doi.org/10.3390/microorganisms13020315 - 1 Feb 2025
Cited by 1 | Viewed by 2123
Abstract
The human microbiome, which encompasses microbial communities and their genetic material, significantly influences health and disease, including cancer. The urogenital microbiota, naturally present in the urinary and genital tracts, interact with factors such as age, lifestyle, and health conditions to affect homeostasis and [...] Read more.
The human microbiome, which encompasses microbial communities and their genetic material, significantly influences health and disease, including cancer. The urogenital microbiota, naturally present in the urinary and genital tracts, interact with factors such as age, lifestyle, and health conditions to affect homeostasis and carcinogenesis. Studies suggest that alterations in this microbiota contribute to the development and progression of genitourinary cancers, emphasizing the concept of oncobiome, which refers to microbial genetic contributions to cancer. Similarly, gut microbiota can influence hormone levels and systemic inflammation, impacting cancers such as cervical and prostate cancer. Advanced studies indicate that microbial communities in genitourinary cancers have distinct profiles that may serve as diagnostic biomarkers or therapeutic targets. Dysbiosis of the urinary microbiota correlates with bladder and kidney cancer. Additionally, gut microbiota influence the effectiveness of cancer treatments. However, further research is necessary to clarify causality, the role of microbial metabolites, and hormonal regulation. The aim of this review is to understand that these dynamics present opportunities for innovative cancer diagnostics and therapies, highlighting the need for integration of microbiology, oncology, and genomics to explore the role of microbiota in genitourinary cancers. For this, a comprehensive search of relevant databases was conducted, applying specific inclusion and exclusion criteria to identify studies examining the association between microbiota and urogenital cancers. Research into the mechanisms by which microbiota influence urogenital cancers may pave the way for new diagnostic and therapeutic approaches, ultimately improving patient outcomes. Full article
(This article belongs to the Special Issue Microbiota in Human Health and Disease)
Show Figures

Figure 1

24 pages, 1668 KiB  
Review
Using New Technologies to Analyze Gut Microbiota and Predict Cancer Risk
by Mohammad Amin Hemmati, Marzieh Monemi, Shima Asli, Sina Mohammadi, Behina Foroozanmehr, Dariush Haghmorad, Valentyn Oksenych and Majid Eslami
Cells 2024, 13(23), 1987; https://doi.org/10.3390/cells13231987 - 1 Dec 2024
Cited by 17 | Viewed by 2846
Abstract
The gut microbiota significantly impacts human health, influencing metabolism, immunological responses, and disease prevention. Dysbiosis, or microbial imbalance, is linked to various diseases, including cancer. It is crucial to preserve a healthy microbiome since pathogenic bacteria, such as Escherichia coli and Fusobacterium nucleatum [...] Read more.
The gut microbiota significantly impacts human health, influencing metabolism, immunological responses, and disease prevention. Dysbiosis, or microbial imbalance, is linked to various diseases, including cancer. It is crucial to preserve a healthy microbiome since pathogenic bacteria, such as Escherichia coli and Fusobacterium nucleatum, can cause inflammation and cancer. These pathways can lead to the formation of tumors. Recent advancements in high-throughput sequencing, metagenomics, and machine learning have revolutionized our understanding of the role of gut microbiota in cancer risk prediction. Early detection is made easier by machine learning algorithms that improve the categorization of cancer kinds based on microbiological data. Additionally, the investigation of the microbiome has been transformed by next-generation sequencing (NGS), which has made it possible to fully profile both cultivable and non-cultivable bacteria and to understand their roles in connection with cancer. Among the uses of NGS are the detection of microbial fingerprints connected to treatment results and the investigation of metabolic pathways implicated in the development of cancer. The combination of NGS with machine learning opens up new possibilities for creating customized medicine by enabling the development of diagnostic tools and treatments that are specific to each patient’s microbiome profile, even in the face of obstacles like data complexity. Multi-omics studies reveal microbial interactions, biomarkers for cancer detection, and gut microbiota’s impact on cancer progression, underscoring the need for further research on microbiome-based cancer prevention and therapy. Full article
Show Figures

Figure 1

19 pages, 1344 KiB  
Review
Microbiota and Autism: A Review on Oral and Gut Microbiome Analysis Through 16S rRNA Sequencing
by Federico Anaclerio, Maria Minelli, Ivana Antonucci, Valentina Gatta and Liborio Stuppia
Biomedicines 2024, 12(12), 2686; https://doi.org/10.3390/biomedicines12122686 - 25 Nov 2024
Cited by 3 | Viewed by 3653
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition with multifactorial etiologies, including genetic, environmental, and microbiological factors. In recent years, increasing attention has been given to the role of the gut microbiota in ASD. Emerging evidence suggests that gut microbiota dysbiosis may [...] Read more.
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition with multifactorial etiologies, including genetic, environmental, and microbiological factors. In recent years, increasing attention has been given to the role of the gut microbiota in ASD. Emerging evidence suggests that gut microbiota dysbiosis may influence the central nervous system through the gut-brain axis, potentially impacting behavior and neurodevelopment. The use of 16S rRNA gene sequencing has become a pivotal tool in profiling the microbial communities associated with ASD, offering valuable insights into bacterial diversity, composition, and potential functional roles. This review aims to provide a comprehensive analysis of current findings on the relationship between the gut and oral microbiota with ASD, and a particular focus on studies utilizing 16S rRNA sequencing. We will explore how gut microbiome alterations may contribute to ASD pathophysiology, discuss the limitations of existing research, and propose future directions for the integration of microbiome analysis in ASD diagnostics and treatment strategies. These findings underscore the potential role of microbiota in modulating ASD symptoms. The data suggest that specific bacterial taxa are consistently altered in ASD, which may have implications for understanding the gut-brain axis and its influence on neurodevelopment. Full article
(This article belongs to the Section Microbiology in Human Health and Disease)
Show Figures

Figure 1

11 pages, 514 KiB  
Protocol
Assessing Diagnostic Performance of Molecular Culture for Neonatal Sepsis: Protocol of the CHAMPIONS Study
by Jip Groen, Martijn van der Kuip, Dries Budding, Martine P. Bos, Marc A. Benninga, Hendrik J. Niemarkt and Tim G. J. de Meij
Diagnostics 2024, 14(17), 1930; https://doi.org/10.3390/diagnostics14171930 - 1 Sep 2024
Viewed by 1772
Abstract
Managing neonatal sepsis is challenging due to nonspecific clinical signs, hematological markers with poor accuracy, and a lengthy turnaround time for the identification of microorganisms. Delaying the initiation of antibiotics in truly infected infants can lead to severe morbidity and mortality. Therefore, decisions [...] Read more.
Managing neonatal sepsis is challenging due to nonspecific clinical signs, hematological markers with poor accuracy, and a lengthy turnaround time for the identification of microorganisms. Delaying the initiation of antibiotics in truly infected infants can lead to severe morbidity and mortality. Therefore, decisions regarding empiric antibiotic treatment are risk stratified, which exposes many uninfected infants to antibiotics. This causes gut microbiota perturbation, unnecessary hospital admissions, and the generation of multi-resistant organisms. High-speed diagnostic assays could expedite discontinuation or avert the initiation of antibiotics in uninfected infants. This study will evaluate the diagnostic performance of molecular culture (MC), a rapid broad-range PCR-based bacterial profiling technique, for diagnosing neonatal sepsis in infants below 90 days old. A multi-center prospective observational cohort study will include infants evaluated for early and late-onset sepsis. Routine evaluation for suspected sepsis includes microbiological cultures of blood. Additionally, blood for MC will be collected. For early-onset sepsis, umbilical cord blood may be used alternatively. Primary outcome is the agreement between MC and conventional blood culture results. Secondary outcome is the agreement of both assays with clinical sepsis using four different, commonly used definitions. Faster diagnostic pathways for sepsis may reduce antibiotic exposure time. Broad-range molecular assays may identify pathogens undetectable by conventional methods. Employment of umbilical cord blood samples for early-onset sepsis diagnosis can resolve challenges in collecting adequate blood volume and could further expedite treatment decisions. Full article
(This article belongs to the Section Diagnostic Microbiology and Infectious Disease)
Show Figures

Figure 1

14 pages, 1644 KiB  
Article
Developing a Symbiotic Fermented Milk Product with Microwave-Treated Hawthorn Extract
by Aidana Utebaeva, Eleonora Gabrilyants and Zhansaya Abish
Fermentation 2024, 10(8), 377; https://doi.org/10.3390/fermentation10080377 - 24 Jul 2024
Cited by 3 | Viewed by 1853
Abstract
The rising interest in functional foods has increased the use of probiotics and prebiotics in fermented dairy products to enhance gut health. This study focuses on developing a symbiotic fermented milk product using Lactobacillus acidophilus and Bifidobacterium bifidum activated with hawthorn extract as [...] Read more.
The rising interest in functional foods has increased the use of probiotics and prebiotics in fermented dairy products to enhance gut health. This study focuses on developing a symbiotic fermented milk product using Lactobacillus acidophilus and Bifidobacterium bifidum activated with hawthorn extract as a prebiotic. Three versions of the product were tested: a control and two variants with B. bifidum activated with 10−5 g/cm3 and 10−10 g/cm3 hawthorn extract, respectively. Key characteristics such as microbiological safety, sensory properties, amino acid profile, vitamin and mineral content, antioxidant capacity, and nutritional values were evaluated. Results showed that products enriched with hawthorn extract had favorable sensory properties and sustained high levels of lactic acid bacteria while being free of pathogens. Product 1 based on L. acidophilus and enriched with B. bifidum activated with hawthorn extract at a concentration of 10−5 g/cm3 demonstrated significant increases in L. acidophilus (24.1%) and B. bifidum (14.7%) after 7 days compared to the control. Both enriched products exhibited slower titratable acidity increases and higher viscosities over 14 days, indicating better preservation and texture stability. Product 1 was notably enriched with essential amino acids, vitamins, and minerals, alongside enhanced antioxidant properties due to increased flavonoid content. The technology developed ensures probiotic viability at 109–1010 CFU/cm3 after 14 days, making it viable for dairy production. Full article
(This article belongs to the Special Issue Analysis of Quality and Sensory Characteristics of Fermented Products)
Show Figures

Figure 1

18 pages, 1020 KiB  
Review
Exploring the Gut Microbiome’s Role in Inflammatory Bowel Disease: Insights and Interventions
by Despoina Gyriki, Christos Nikolaidis, Elisavet Stavropoulou, Ioanna Bezirtzoglou, Christina Tsigalou, Stergios Vradelis and Eugenia Bezirtzoglou
J. Pers. Med. 2024, 14(5), 507; https://doi.org/10.3390/jpm14050507 - 11 May 2024
Cited by 14 | Viewed by 5787
Abstract
Inflammatory Bowel Disease (IBD), encompassing Crohn’s disease (CD) and ulcerative colitis (UC), is a chronic and relapsing inflammatory condition of the intestine that significantly impairs quality of life and imposes a heavy burden on healthcare systems globally. While the exact etiology of IBD [...] Read more.
Inflammatory Bowel Disease (IBD), encompassing Crohn’s disease (CD) and ulcerative colitis (UC), is a chronic and relapsing inflammatory condition of the intestine that significantly impairs quality of life and imposes a heavy burden on healthcare systems globally. While the exact etiology of IBD is unclear, it is influenced by genetic, environmental, immunological, and microbial factors. Recent advances highlight the gut microbiome’s pivotal role in IBD pathogenesis. The microbial dysbiosis characteristic of IBD, marked by a decline in beneficial bacteria and an increase in pathogenic microbes, suggests a profound connection between microbial imbalance and disease mechanisms. This review explores diagnostic approaches to IBD that integrate clinical assessment with advanced microbiological analyses, highlighting the potential of microbiome profiling as a non-invasive diagnostic tool. In addition, it evaluates conventional and emerging treatments and discusses microbiome-targeted intervention prospects, such as probiotics, symbiotics, and faecal microbiota transplantation. The necessity for future research to establish their efficacy and safety is emphasised. Full article
(This article belongs to the Special Issue Gut Microbiome and Its Impact on Human Health)
Show Figures

Figure 1

18 pages, 8459 KiB  
Article
Mulberry Leaf Dietary Supplementation Can Improve the Lipo-Nutritional Quality of Pork and Regulate Gut Microbiota in Pigs: A Comprehensive Multi-Omics Analysis
by Junjie Hou, Xiang Ji, Xiaoran Chu, Binjie Wang, Kangle Sun, Haibo Wei, Yu Zhang, Zhen Song and Fengyun Wen
Animals 2024, 14(8), 1233; https://doi.org/10.3390/ani14081233 - 19 Apr 2024
Cited by 2 | Viewed by 2235
Abstract
Mulberry leaves, a common traditional Chinese medicine, represent a potential nutritional strategy to improve the fat profile, also known as the lipo-nutrition, of pork. However, the effects of mulberry leaves on pork lipo-nutrition and the microorganisms and metabolites in the porcine gut remain [...] Read more.
Mulberry leaves, a common traditional Chinese medicine, represent a potential nutritional strategy to improve the fat profile, also known as the lipo-nutrition, of pork. However, the effects of mulberry leaves on pork lipo-nutrition and the microorganisms and metabolites in the porcine gut remain unclear. In this study, multi-omics analysis was employed in a Yuxi black pig animal model to explore the possible regulatory mechanism of mulberry leaves on pork quality. Sixty Yuxi black pigs were divided into two groups: the control group (n = 15) was fed a standard diet, and the experimental group (n = 45) was fed a diet supplemented with 8% mulberry leaves. Experiments were performed in three replicates (n = 15 per replicate); the two diets were ensured to be nutritionally balanced, and the feeding period was 120 days. The results showed that pigs receiving the diet supplemented with mulberry leaves had significantly reduced backfat thickness (p < 0.05) and increased intramuscular fat (IMF) content (p < 0.05) compared with pigs receiving the standard diet. Lipidomics analysis showed that mulberry leaves improved the lipid profile composition and increased the proportion of triglycerides (TGs). Interestingly, the IMF content was positively correlated with acyl C18:2 and negatively correlated with C18:1 of differential TGs. In addition, the cecal microbiological analysis showed that mulberry leaves could increase the abundance of bacteria such as UCG-005, Muribaculaceae_norank, Prevotellaceae_NK3B31_group, and Limosilactobacillus. Simultaneously, the relative levels of L-tyrosine-ethyl ester, oleic acid methyl ester, 21-deoxycortisol, N-acetyldihydrosphingosine, and mulberrin were increased. Furthermore, we found that mulberry leaf supplementation significantly increased the mRNA expression of lipoprotein lipase, fatty acid-binding protein 4, and peroxisome proliferators-activated receptor γ in muscle (p < 0.01). Mulberry leaf supplementation significantly increased the mRNA expression of diacylglycerol acyltransferase 1 (p < 0.05) while significantly decreasing the expression of acetyl CoA carboxylase in backfat (p < 0.05). Furthermore, mulberry leaf supplementation significantly upregulated the mRNA expression of hormone-sensitive triglyceride lipase and peroxisome proliferator-activated receptor α (p < 0.05) in backfat. In addition, mulberry leaf supplementation led to increased serum leptin and adiponectin (p < 0.01). Collectively, this omic profile is consistent with an increased ratio of IMF to backfat in the pig model. Full article
(This article belongs to the Section Animal Nutrition)
Show Figures

Figure 1

17 pages, 4253 KiB  
Article
GroEL-Proteotyping of Bacterial Communities Using Tandem Mass Spectrometry
by Simon Klaes, Shobhit Madan, Darja Deobald, Myriel Cooper and Lorenz Adrian
Int. J. Mol. Sci. 2023, 24(21), 15692; https://doi.org/10.3390/ijms242115692 - 28 Oct 2023
Cited by 2 | Viewed by 2476
Abstract
Profiling bacterial populations in mixed communities is a common task in microbiology. Sequencing of 16S small subunit ribosomal-RNA (16S rRNA) gene amplicons is a widely accepted and functional approach but relies on amplification primers and cannot quantify isotope incorporation. Tandem mass [...] Read more.
Profiling bacterial populations in mixed communities is a common task in microbiology. Sequencing of 16S small subunit ribosomal-RNA (16S rRNA) gene amplicons is a widely accepted and functional approach but relies on amplification primers and cannot quantify isotope incorporation. Tandem mass spectrometry proteotyping is an effective alternative for taxonomically profiling microorganisms. We suggest that targeted proteotyping approaches can complement traditional population analyses. Therefore, we describe an approach to assess bacterial community compositions at the family level using the taxonomic marker protein GroEL, which is ubiquitously found in bacteria, except a few obligate intracellular species. We refer to our method as GroEL-proteotyping. GroEL-proteotyping is based on high-resolution tandem mass spectrometry of GroEL peptides and identification of GroEL-derived taxa via a Galaxy workflow and a subsequent Python-based analysis script. Its advantage is that it can be performed with a curated and extendable sample-independent database and that GroEL can be pre-separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) to reduce sample complexity, improving GroEL identification while simultaneously decreasing the instrument time. GroEL-proteotyping was validated by employing it on a comprehensive raw dataset obtained through a metaproteome approach from synthetic microbial communities as well as real human gut samples. Our data show that GroEL-proteotyping enables fast and straightforward profiling of highly abundant taxa in bacterial communities at reasonable taxonomic resolution. Full article
(This article belongs to the Special Issue Advances in Mass Spectrometry-Based Proteomics)
Show Figures

Figure 1

14 pages, 323 KiB  
Article
Effect of Lactobacillus kefiri, in Conjunction with PENS T6 and a Hypocaloric Diet, on Weight Loss, Hypertension and Laboratory Glycemic and Lipid Profile
by Jaime Ruiz-Tovar, Carolina Llavero and Maria-Encarnacion Fernandez-Contreras
Nutrients 2023, 15(21), 4549; https://doi.org/10.3390/nu15214549 - 26 Oct 2023
Cited by 3 | Viewed by 1925
Abstract
The pathogenesis of obesity has been linked to alterations in gut microorganisms. The aim of this study was to investigate the effect of Lactobacillus kefiri, together with PENS T6 and a hypocaloric diet, on weight loss, hypertension and laboratory glycemic and lipid [...] Read more.
The pathogenesis of obesity has been linked to alterations in gut microorganisms. The aim of this study was to investigate the effect of Lactobacillus kefiri, together with PENS T6 and a hypocaloric diet, on weight loss, hypertension and laboratory glycemic and lipid profile. A prospective non-randomized study was conducted involving adult patients with a body mass index (BMI) > 30 kg/m2. Patients were divided into two groups: those undergoing PENS-T6 and hypocaloric diet (PENS-Diet Group) and those undergoing the same PENS-T6 scheme and hypocaloric diet, but additionally receiving probiotics including Lactobacillus kefiri (PENS-Diet + L. kefiri Group). Weight loss was assessed at the end of the treatment, and analytical glycemic and lipid profile, and microbiological analysis of feces were performed before and after treatment. The addition of Lactobacillus kefiri to PENS T6 and a low-calorie diet, increases weight loss and further improves the glycemic and lipid profile. L. kefiri also causes a further improvement in obesity-associated dysbiosis, mainly by increasing the muconutritive (Akkermansia muciniphila) and regulatory (Bifidobacterium spp.) microbiome, and the Phylum Bacteroidetes (Prevotella spp.) and decreasing the Firmicutes/Bacteroidetes ratio. Full article
(This article belongs to the Section Prebiotics and Probiotics)
13 pages, 1155 KiB  
Article
The Modulation Effect of a Fermented Bee Pollen Postbiotic on Cardiovascular Microbiota and Therapeutic Perspectives
by Laura-Dorina Dinu, Florentina Gatea, Florentina Roaming Israel, Milena Lakicevic, Nebojša Dedović and Emanuel Vamanu
Biomedicines 2023, 11(10), 2712; https://doi.org/10.3390/biomedicines11102712 - 5 Oct 2023
Cited by 12 | Viewed by 3221
Abstract
Hypertension is a frequent comorbidity in patients with heart failure; therefore, blood pressure management for these patients is widely recommended in medical guidelines. Bee pollen and postbiotics that contain inactivated probiotic cells and their metabolites have emerged as promising bioactive compounds sources, and [...] Read more.
Hypertension is a frequent comorbidity in patients with heart failure; therefore, blood pressure management for these patients is widely recommended in medical guidelines. Bee pollen and postbiotics that contain inactivated probiotic cells and their metabolites have emerged as promising bioactive compounds sources, and their potential role in mitigating cardiovascular (CV) risks is currently being unveiled. Therefore, this preliminary study aimed to investigate the impact of a lactic-fermented bee pollen postbiotic (FBPP) on the CV microbiota via in vitro tests. A new isolated Lactobacillus spp. strain from the digestive tract of bees was used to ferment pollen, obtaining liquid and dried atomized caps postbiotics. The modulating effects on a CV microbiota that corresponds to the pathophysiology of hypertension were investigated using microbiological methods and qPCR and correlated with the metabolic profile. Both liquid and dried FBPPs increased the number of the beneficial Lactobacillus spp. and Bifidobacterium spp. bacteria by up to 2 log/mL, while the opportunistic pathogen E. coli, which contributes to CV pathogenesis, decreased by 3 log/mL. The short-chain fatty acid (SCFA) profile revealed a significant increase in lactic (6.386 ± 0.106 g/L) and acetic (4.284 ± 0.017 g/L) acids, both with known antihypertensive effects, and the presence of isovaleric acid, which promotes a healthy gut microbiota. Understanding the impact of the FBPP on gut microbiota could lead to innovative strategies for promoting heart health and preventing cardiovascular diseases. Full article
(This article belongs to the Special Issue Recent Advances in Gut Microbiome and Heart Failure)
Show Figures

Figure 1

17 pages, 2223 KiB  
Article
Bariatric Surgery as Treatment Strategy of Obesity in Saudi People: Effects of Gut Microbiota
by Seham J. Alqahtani, Hanan A. Alfawaz, Nadine M. S. Moubayed, Wail M. Hassan, Ahmad T. Almnaizel, Noura M. S. Alshiban, Jawahir M. Abuhaimed, Mohammed F. Alahmed, Mosffer M. AL-Dagal and Afaf El-Ansary
Nutrients 2023, 15(2), 361; https://doi.org/10.3390/nu15020361 - 11 Jan 2023
Cited by 8 | Viewed by 3765
Abstract
Obesity prevalence is rising globally, as are the number of chronic disorders connected with obesity, such as diabetes, non-alcoholic fatty liver disease, dyslipidemia, and hypertension. Bariatric surgery is also becoming more common, and it remains the most effective and long-term treatment for obesity. [...] Read more.
Obesity prevalence is rising globally, as are the number of chronic disorders connected with obesity, such as diabetes, non-alcoholic fatty liver disease, dyslipidemia, and hypertension. Bariatric surgery is also becoming more common, and it remains the most effective and long-term treatment for obesity. This study will assess the influence of Laparoscopic Sleeve Gastrectomy (LSG) on gut microbiota in people with obesity before and after surgery. The findings shed new light on the changes in gut microbiota in Saudi people with obesity following LSG. In conclusion, LSG may improve the metabolic profile, resulting in decreased fat mass and increased lean mass, as well as improving the microbial composition balance in the gastrointestinal tract, but this is still not equivalent to normal weight microbiology. A range of factors, including patient characteristics, geographic dispersion, type of operation, technique, and nutritional and caloric restriction, could explain differences in abundance between studies. This information could point to a novel and, most likely, tailored strategy in obesity therapy, which could eventually be incorporated into health evaluations and monitoring in preventive health care or clinical medicine. Full article
(This article belongs to the Section Nutrition and Obesity)
Show Figures

Figure 1

Back to TopTop