Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.445
Filtrar
1.
Oncol Ther ; 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-39217582

RESUMEN

The gut microbiota plays a crucial role in maintaining homeostasis in the human gastrointestinal tract. Numerous studies have shown a strong association between the gut microbiota and the emergence and progression of various diseases. Leukemia is one of the most common hematologic malignancies. Although standardized protocols and expert consensus have been developed for routine diagnosis and treatment, limitations remain due to individual differences. Nevertheless, a large number of studies have established a link between the gut microbiota and leukemia, with disturbances in the gut microbiota directly or indirectly affecting the development of leukemia. However, the causal relationship between the two remains unclear, and studying and exploring the causal relationship may open up entirely new avenues and protocols for use in the prevention and/or treatment of leukemia, offering new insights into diagnosis and treatment. In this review, the intricate relationship between the gut microbiota and leukemia is explored in depth, including causal associations, metabolite effects, therapeutic applications, and complications. Based on the characteristics of the gut microbiota, the future applications and prospects of gut microbiota are discussed to provide useful information for clinical treatment of leukemia.

2.
J Nutr Sci Vitaminol (Tokyo) ; 70(4): 311-317, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39218692

RESUMEN

Chronic inflammation in adipose tissue is thought to contribute to insulin resistance, which involves the gut microbiota. Our previous studies have demonstrated that ingestion of 1-kestose can alter the gut microbiota composition, increase cecal butyrate levels, and improve insulin resistance in Otsuka Long-Evans Tokushima Fatty (OLETF) rats. Additionally, we found that 1-kestose supplementation ameliorated insulin resistance in obese rat models fed a high-fat diet (HFD), although the effects of 1-kestose on the abundance of inflammation-related gene in adipose tissue and gut microbiota composition in these rats were not explored. This study aimed to investigate the impact of 1-kestose on these parameters in HFD-fed rats, compared to OLETF rats. Male Sprague-Dawley rats were divided into two dietary groups, control or HFD, for 19 wk. Each group was further subdivided to receive either tap water or tap water supplemented with 2% (w/v) 1-kestose throughout the study. We evaluated gene expression in adipose tissue, as well as short-chain fatty acids (SCFAs) levels and microbial composition in the cecum contents. 1-Kestose intake restored the increased relative abundance of tumor necrosis factor (Tnf) mRNA in adipose tissue and the reduced level of butyrate in the cecum contents of HFD-fed rats to those observed in control diet-fed rats. Additionally, 1-kestose consumption changed the composition of the gut microbiota, increasing Butyricicoccus spp., decreasing UGC-005 and Streptococcus spp., in the cecum contents of HFD-fed rats. Our findings suggest that 1-kestose supplementation reduces adipose tissue inflammation and increases butyrate levels in the gut of HFD-fed rats, associated with changes in the gut microbiota composition, distinct from those seen in OLETF rats.


Asunto(s)
Tejido Adiposo , Ciego , Dieta Alta en Grasa , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Inflamación , ARN Mensajero , Ratas Sprague-Dawley , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Masculino , Tejido Adiposo/metabolismo , Tejido Adiposo/efectos de los fármacos , Inflamación/metabolismo , ARN Mensajero/metabolismo , Ratas , Ácidos Grasos Volátiles/metabolismo , Ciego/microbiología , Ciego/metabolismo , Resistencia a la Insulina , Ratas Endogámicas OLETF , Obesidad/metabolismo , Obesidad/microbiología , Suplementos Dietéticos , Butiratos/metabolismo
3.
Pediatr Allergy Immunol ; 35(9): e14226, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39221598

RESUMEN

BACKGROUND: Immunomodulatory proteins in human milk (HM) can shape infant immune development. However, strategies to modulate their levels are currently unknown. This study investigated whether maternal prebiotic supplementation alters the levels of immunomodulatory proteins in HM. METHODS: The study was nested within the SYMBA double-blind randomized controlled trial (ACTRN12615001075572), which investigated the effects of maternal prebiotic (short-chain galacto-oligosaccharides/long-chain fructo-oligosaccharides) supplementation from <21 weeks gestation during pregnancy until 6 months postnatal during lactation on child allergic disease risk. Mother-child dyads receiving prebiotics (n = 46) or placebo (n = 54) were included in this study. We measured the levels of 24 immunomodulatory proteins in HM collected at 2, 4, and 6 months. RESULTS: Cluster analysis showed that the overall immunomodulatory protein composition of milk samples from both groups was similar. At 2 months, HM of prebiotic-supplemented women had decreased levels of TGF-ß1 and TSLP (95% CI: -17.4 [-29.68, -2.28] and -57.32 [-94.22, -4.7] respectively) and increased levels of sCD14 (95% CI: 1.81 [0.17, 3.71]), when compared to the placebo group. At 4 months, IgG1 was lower in the prebiotic group (95% CI: -1.55 [-3.55, -0.12]) compared to placebo group. CONCLUSION: This exploratory study shows that prebiotic consumption by lactating mothers selectively alters specific immunomodulatory proteins in HM. This finding is crucial for understanding how prebiotic dietary recommendations for pregnant and lactating women can modify the immune properties of HM and potentially influence infant health outcomes through immune support from breastfeeding.


Asunto(s)
Suplementos Dietéticos , Leche Humana , Prebióticos , Humanos , Leche Humana/inmunología , Leche Humana/química , Prebióticos/administración & dosificación , Femenino , Método Doble Ciego , Embarazo , Lactante , Adulto , Masculino , Lactancia/inmunología , Oligosacáridos/administración & dosificación , Recién Nacido , Lactancia Materna , Citocinas/metabolismo
4.
Brain Behav Immun ; 122: 167-184, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-39142421

RESUMEN

Ageing is characterised by a progressive increase in systemic inflammation and especially neuroinflammation. Neuroinflammation is associated with altered brain states that affect behaviour, such as an increased level of anxiety with a concomitant decline in cognitive abilities. Although multiple factors play a role in the development of neuroinflammation, microglia have emerged as a crucial target. Microglia are the only macrophage population in the CNS parenchyma that plays a crucial role in maintaining homeostasis and in the immune response, which depends on the activation and subsequent deactivation of microglia. Therefore, microglial dysfunction has a major impact on neuroinflammation. The gut microbiota has been shown to significantly influence microglia from birth to adulthood in terms of development, proliferation, and function. Diet is a key modulating factor that influences the composition of the gut microbiota, along with prebiotics that support the growth of beneficial gut bacteria. Although the role of diet in neuroinflammation and behaviour has been well established, its relationship with microglia functionality is less explored. This article establishes a link between diet, animal behaviour and the functionality of microglia. The results of this research stem from experiments on mouse behaviour, i.e., memory, anxiety, and studies on microglia functionality, i.e., cytochemistry (phagocytosis, cellular senescence, and ROS assays), gene expression and protein quantification. In addition, shotgun sequencing was performed to identify specific bacterial families that may play a crucial role in the brain function. The results showed negative effects of long-term consumption of a high fat diet on ageing mice, epitomised by increased body weight, glucose intolerance, anxiety, cognitive impairment and microglia dysfunction compared to ageing mice on a control diet. These effects were a consequence of the changes in gut microbiota modulated by the diet. However, by adding the prebiotics fructo- and galacto-oligosaccharides, we were able to mitigate the deleterious effects of a long-term high-fat diet.

5.
Cureus ; 16(8): e66994, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39161553

RESUMEN

BACKGROUND AND AIMS: The use of gut biotics, including probiotics, prebiotics, and synbiotics, has shown substantial potential in the management of various health conditions possibly through the gut-organ axis. The role of gut biotics in modulating the gut-brain axis is becoming evident with more research focusing on this intervention. Improvement of gut-organ axis function is possible by using food-related products called gut biotics. However, there is limited comprehension of the knowledge and use of these intestinal or gut biotics. Our aim was to recognize knowledge gaps and assess the improvement of understanding following an education intervention. METHODS: A single-arm study encompassing a convenient sample of 161 inpatient and outpatient subjects aged 50 years and older was conducted at the University of Alberta Hospital from June to August 2023. Knowledge about gut biotics was evaluated using a structured questionnaire consisting of 16 questions and involving six thematic areas. To ensure validity, the questionnaire was pre-tested on 10 physicians and residents who were not part of the study. The questionnaire was administered to study subjects prior to receiving an information sheet about gut biotics. Two weeks after receiving the information sheet, all participants were contacted by phone, and the same questionnaire was administered again. Of the 161 patients, 122 completed the pre-intervention and post-intervention questionnaires and were considered in the analysis. RESULTS: The mean age of the participants was 72 years (SD: 10.8), 57% comprised women, and 39% had less than a high school education. The proportion of polypharmacy and multimorbidity was 87% and 97%, respectively. Following the intervention, there was a noticeable enhancement in knowledge across all the themes, with statistical significance (p<0.001) observed in 14 out of 16 questions as determined by the homogeneity statistical test. CONCLUSIONS: Knowledge gaps in gut biotics were prevalent among study participants, and the educational intervention effectively contributed to the enhancement of knowledge. The results of this study provide valuable information for the development of targeted health education strategies focusing on gut biotics, which may play a role in improving gut-organ axis function.

6.
Gut Microbes ; 16(1): 2392009, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39161102

RESUMEN

Here, we explored the vast potential of microbiome-based interventions in preventing and managing non-communicable diseases including obesity, diabetes, allergies, celiac disease, inflammatory bowel diseases, malnutrition, and cardiovascular diseases across different life stages. We discuss the intricate relationship between microbiome and non-communicable diseases, emphasizing on the "window of opportunity" for microbe-host interactions during the first years after birth. Specific biotics and also live biotherapeutics including fecal microbiota transplantation emerge as pivotal tools for precision medicine, acknowledging the "one size doesn't' fit all" aspect. Challenges in implementation underscore the need for advanced technologies, scientific transparency, and public engagement. Future perspectives advocate for understanding maternal-neonatal microbiome, exploring the maternal exposome and delving into human milk's role in the establishment and restoration of the infant microbiome and its influence over health and disease. An integrated scientific approach, employing multi-omics and accounting for inter-individual variance in microbiome composition and function appears central to unleash the full potential of early-life microbiome interventions in revolutionizing healthcare.


Asunto(s)
Microbioma Gastrointestinal , Humanos , Embarazo , Femenino , Recién Nacido , Leche Humana/microbiología , Trasplante de Microbiota Fecal , Lactante , Interacciones Microbiota-Huesped
8.
Front Nutr ; 11: 1403007, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39183984

RESUMEN

Prebiotics can modulate the gut microbial community composition and function for improved (gut) health and increase resilience against infections. In vitro models of the gut facilitate the study of intervention effects on the gut microbial community relevant to health. The mucosa-associated gut microbiota, which thrives in close contact with the host plays a pivotal role in colonization resistance and health. Therefore, we here introduce the Mi-screen, an experimental approach implementing a 96-well plate equipped with a mucus agar layer for the additional culturing of mucosa-associated microbiota in vitro. In this study, we screened the effects of 2'-Fucosyllactose (2'-FL), fructooligosaccharides (FOS), and inulin within a complex microbiota without and with infection with the C. difficile strains ATCC 43599 (Ribotype 001) or ATCC BAA-1870 (Ribotype 027). We analyzed the microbial community composition and short-chain fatty acid levels after 48 h of incubation. The inclusion of an additional substrate and surface in the form of the mucus agar layer allowed us to culture a microbial richness ranging between 100-160 in Chao index, with Shannon indices of 5-6 across culture conditions, indicative of a microbial diversity of physiological relevance. The mucus agar layer stimulated the growth of characteristic mucosa-associated bacteria such as Roseburia inulinovorans. The prebiotic interventions affected luminal and mucosal microbial communities cultured in vitro and stimulated short-chain fatty acid production. FOS, inulin and 2'-FL promoted the growth of Bifidobacterium adolescentis within the mucosa-associated microbiota cultured in vitro. When spiking the untreated conditions with pathogenic C. difficile, the strains thrived within the luminal and the mucosal sample types, whereas prebiotic treatments exhibited inhibitory effects on C. difficile growth and prevented colonization. In conclusion, the Mi-screen facilitates the screening of luminal and mucosa-associated gut microbial community dynamics in vitro and therefore fills an important gap in the field of in vitro modeling.

9.
Transl Anim Sci ; 8: txae104, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39185353

RESUMEN

The objective of this study was to evaluate the effects of yeast products (YP) and essential oils (EO) in total or partial replacement to in-feed antibiotic protocols (growth promoter and prophylactic), both in recommended doses and in overdose of prophylactic antibiotics (PA), on growth performance, and diarrhea incidence in the growing-finishing pigs; and fecal microbiota in market hogs. Four hundred pigs (20.36 ±â€…2.64 kg) were assigned to five treatments in a randomized block design: diets with prophylactic and growth promoter antibiotics (ANT); ANT with 30% more PA (ANT+30); diets with less PA and YP (ANT+Y); diets with less PA, YP and EO (ANT+Y+EO); and antibiotics-free diets with YP and EO (Y+EO). The content of the active components of the YP was 60% purified ß-1,3/1,6-glucans extracted from Saccharomyces cerevisiae yeast (Macrogard), 20% functional water-soluble MOS (HyperGen), and 18% MOS, extracted from Saccharomyces cerevisiae yeast (ActiveMOS). From 0 to 14 d, pigs of the ANT+30, ANT+Y, and ANT+Y+EO treatments showed a greater body weight (BW) and average daily gain (ADG) compared to pigs from the Y+EO group. From 14 to 35 d, pigs of ANT+30 and ANT+Y+EO treatments were heavier than Y+EO group. At 105 d, ANT pigs had a higher BW than the Y+EO group. For the entire period, ADG of ANT pigs was greater, and feed conversion ratio better than Y+EO pigs. From 0 to 35 d, pigs of the Y+EO treatment showed a higher diarrhea incidence compared to pigs of the other groups. From 49 to 70 d, ANT+Y and ANT+Y+EO treatments showed a lower diarrhea incidence than Y+EO group, which remained the case during the overall period. At 105 d, the alpha diversity of fecal microbiota by Shannon Entropy was lower in ANT, ANT+30, and Y+EO groups than observed for ANT+Y+EO group. The abundance of Firmicutes phylum and Firmicutes/Bacteroidetes ratio was higher in ANT than in ANT+Y+EO pigs. Proteobacteria phylum abundance in ANT+Y+EO was higher than ANT, ANT+Y, and Y+EO. Peptostreptococcaceae family abundance was higher in ANT, ANT+30, and ANT+Y groups than in ANT+Y+EO and Y+EO groups. ANT+Y+EO and Y+EO groups show a lower abundance of SMB53 genus than ANT and ANT+30 groups. In conclusion, the use of YP and EO, in partial replacement to the in-feed antibiotic protocols, does not reduce the growth performance, can replace antibiotic growth promotors, and reduce the in-feed use of PA in growing-finishing pigs. The use of YP and EO, together with PA, increases the microbial diversity, despite having important genera for weight gain in less abundance. Overdose of PA does not improve growth performance and reduces microbial diversity, which does not characterize it as an efficient preventive protocol.

10.
Front Nutr ; 11: 1411374, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39171106

RESUMEN

Childhood obesity presents a serious health concern associated with gut microbiota alterations. Dietary interventions targeting the gut microbiota have emerged as promising strategies for managing obesity in children. This study aimed to elucidate the impact of stachyose (STS) supplementation on the gut microbiota composition and metabolic processes in obese children. Fecal samples were collected from 40 obese children (20 boys and 20 girls) aged between 6 and 15 and in vitro fermentation was conducted with or without the addition of STS, respectively, followed by 16S rRNA amplicon sequencing and analysis of short-chain fatty acids (SCFAs) and gases. Notably, our results revealed that STS supplementation led to significant alterations in gut microbiota composition, including an increase in the abundance of beneficial bacteria such as Bifidobacterium and Faecalibacterium, and a decrease in harmful bacteria including Escherichia-Shigella, Parabacteroides, Eggerthella, and Flavonifractor. Moreover, STS supplementation resulted in changes in SCFAs production, with significant increases in acetate levels and reductions in propionate and propionate, while simultaneously reducing the generation of gases such as H2S, H2, and NH3. The Area Under the Curve (AUC)-Random Forest algorithm and PICRUSt 2 were employed to identify valuable biomarkers and predict associations between the gut microbiota, metabolites, and metabolic pathways. The results not only contribute to the elucidation of STS's modulatory effects on gut microbiota but also underscore its potential in shaping metabolic activities within the gastrointestinal environment. Furthermore, our study underscores the significance of personalized nutrition interventions, particularly utilizing STS supplementation, in the management of childhood obesity through targeted modulation of gut microbial ecology and metabolic function.

11.
Artículo en Inglés | MEDLINE | ID: mdl-39173718

RESUMEN

BACKGROUND: Ingestion of prebiotics during pregnancy and lactation may have immunomodulatory benefits for the developing fetal and infant immune system and provide a potential dietary strategy to reduce the risk of allergic diseases. OBJECTIVE: The aim of this trial was to determine whether maternal supplementation with dietary prebiotics reduces the risk of allergic outcomes in infants with hereditary risk. METHODS: We undertook a double-blind, randomized controlled trial in which pregnant women were allocated to consume prebiotics (14.2g daily of galacto-oligosaccharides and fructo-oligosaccharides in ratio 9:1) or placebo (8.7g daily maltodextrin) powder from <21 weeks gestation until 6-months postnatal during lactation. Eligible women had infants with a first-degree relative with a history of medically diagnosed allergic disease. The primary outcome was infant medically diagnosed eczema by 1-year of age, and secondary outcomes included allergen sensitization, food allergy, and recurrent wheeze by 1-year of age. RESULTS: 652 women were randomized between June 2016 and November 2021 (n=329 prebiotics, n=323 placebo). There was no significant difference between groups in the percentage of infants with medically diagnosed eczema by 1-year of age (prebiotics 31.5% (103/327 infants) compared to placebo 32.6% (105/322 infants); adjusted relative risk 0.98 (95% CI 0.77, 1.23; p=0.84). Secondary outcomes and safety measures also did not significantly differ between groups. CONCLUSION: We found little evidence that maternal prebiotics supplementation during pregnancy and lactation reduces the risk of infant medically diagnosed eczema by 1-year of age in infants who are at hereditary risk of allergic disease.

12.
J Am Acad Dermatol ; 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39173885

RESUMEN

In Part I of our CME we reviewed the skin microbiome in healthy individuals. Part II reviews the evolving understanding of alterations in the skin microbiome in specific human diseases. We also discuss how the skin microbiome can change with environmental exposures and medications such as antibiotics as well as ongoing research on microbiome-based interventions.

13.
Antibiotics (Basel) ; 13(8)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39200020

RESUMEN

The gut microbiota establishes a mutually beneficial relationship with the host starting from birth, impacting diverse metabolic and immunological processes. Dysbiosis, characterized by an imbalance of microorganisms, is linked to numerous medical conditions, including gastrointestinal disorders, cardiovascular diseases, and autoimmune disorders. This imbalance promotes the proliferation of toxin-producing bacteria, disrupts the host's equilibrium, and initiates inflammation. Genetic factors, dietary choices, and drug use can modify the gut microbiota. However, there is optimism. Several therapeutic approaches, such as probiotics, prebiotics, synbiotics, postbiotics, microbe-derived products, and microbial substrates, aim to alter the microbiome. This review thoroughly explores the therapeutic potential of these microbiota modulators, analysing recent studies to evaluate their efficacy and limitations. It underscores the promise of microbiota-based therapies for treating dysbiosis-related conditions. This article aims to ensure practitioners feel well-informed and up to date on the most influential methods in this evolving field by providing a comprehensive review of current research.

14.
J Parkinsons Dis ; 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39213091

RESUMEN

There is increasing evidence that microbial-based therapies can be useful in people with Parkinson's disease (PD). In this viewpoint, we provide a state-of-the-art review of the clinical and pre-clinical evidence for probiotics and prebiotics in PD. Currently, short-term clinical studies, including double-blind placebo-controlled randomized clinical trials, have demonstrated safety, and efficacy primarily in improving constipation-related symptoms. Pre-clinical studies consistently reported improvements in a range of biological markers and outcomes, including evidence for attenuation of gut dysfunction and neuroprotection. Bacteria from the genus Lactobacillus and Bifidobacterium have been the most frequently studied both in clinical and pre-clinical probiotics studies, while research into prebiotics is still limited and primarily involved resistant starch and fructooligosaccharides. We provide practical suggestions for clinicians on how to advise patients in the clinic regarding these popular treatments, and important caveats to be aware of. Finally, areas for further advancements are highlighted. It is envisaged that in the future, microbial-based therapies may benefit from personalization based on an enhanced understanding of a whole range of host factors and host-microbiome interactions.

15.
Diabetes Metab Syndr ; 18(8): 103110, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39213690

RESUMEN

BACKGROUND: We aimed to explore how probiotics, prebiotics, or synbiotics impact glycemic indices in patients with diabetes mellitus. METHOD: A comprehensive search was conducted on PubMed, Scopus, and Web of Science from inception up to April 2023. The random-effects model was employed for the study analysis. Furthermore, sensitivity and subgroup analyses were conducted to investigate potential sources of heterogeneity. AMSTAR2 checklist was used to determine the quality of studies. Comprehensive meta-analysis version 3 was used for the study analysis. RESULT: A total of 31 studies were included in the final analysis. Based on the results of the meta-analysis, gut microbial therapy could significantly decrease serum fasting blood glucose levels in patients with type 2 diabetes mellitus (effect size: -0.211; 95 % CI: -0.257, -0.164; P < 0.001). Additionally, significant associations were also found between gut microbial therapy and improved serum levels of fasting insulin, glycated hemoglobin, and homeostatic model assessment for insulin resistance (effect size: -0.087; 95 % confidence interval: -0.120, -0.053; P < 0.001; effect size: -0.166; 95 % confidence interval: -0.200, -0.132; P < 0.001; effect size: -0.230; 95 % confidence interval: -0.288, -0.172; P < 0.001, respectively). CONCLUSION: Our results revealed promising effects of gut microbiota modulation on glycemic profile of patients with type 2 diabetes mellitus. The use of these agents as additional treatments can be considered.

16.
Microorganisms ; 12(8)2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39203424

RESUMEN

Probiotics are "live microorganisms which, when administered in adequate amount, confer health benefits on the host". They can be found in certain foods like yogurt and kefir and in dietary supplements. The introduction of bacterial derivatives has not only contributed to disease control but has also exhibited promising outcomes, such as improved survival rates, immune enhancement, and growth promotion effects. It is interesting to note that the efficacy of probiotics goes beyond the viability of the bacteria, giving rise to concepts like paraprobiotics, non-viable forms of probiotics, and postbiotics. Paraprobiotics offer various health benefits in children with intestinal dysbiosis, contributing to improved digestive health, immune function, and overall well-being. In this review, the potential of these therapeutic applications as alternatives to pharmacological agents for treating pediatric intestinal dysbiosis will be thoroughly evaluated. This includes an analysis of their efficacy, safety, long-term benefits, and their ability to restore gut microbiota balance, improve digestive health, enhance immune function, and reduce inflammation. The aim is to determine if these non-pharmacological interventions can effectively and safely manage intestinal dysbiosis in children, reducing the need for conventional medications and their side effects.

17.
Microorganisms ; 12(8)2024 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-39203553

RESUMEN

Effects of pre- and probiotics on intestinal health are well researched and microbiome-targeting solutions are commercially available. Even though a trend to appreciate the presence of certain microbes on the skin is seeing an increase in momentum, our understanding is limited as to whether the utilization of skin-resident microbes for beneficial effects holds the same potential as the targeted manipulation of the gut microflora. Here, we present a selection of molecular mechanisms of cross-communication between human skin and the skin microbial community and the impact of these interactions on the host's cutaneous health with implications for the development of skin cosmetic and therapeutic solutions. Malassezia yeasts, as the main fungal representatives of the skin microfloral community, interact with the human host skin via lipid mediators, of which several are characterized by exhibiting potent anti-inflammatory activities. This review therefore puts a spotlight on Malassezia and provides a comprehensive overview of the current state of knowledge about these fungal-derived lipid mediators and their capability to reduce aesthetical and sensory burdens, such as redness and itching, commonly associated with inflammatory skin conditions. Finally, several examples of current skin microbiome-based interventions for cosmetic solutions are discussed, and models are presented for the use of skin-resident microbes as endogenous bio-manufacturing platforms for the in situ supplementation of the skin with beneficial metabolites.

18.
Pathogens ; 13(8)2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-39204246

RESUMEN

Clostridioides difficile is the main causative agent of antibiotic-associated diarrhea (AAD) in hospitals in the developed world. Both infected patients and asymptomatic colonized individuals represent important transmission sources of C. difficile. C. difficile infection (CDI) shows a large range of symptoms, from mild diarrhea to severe manifestations such as pseudomembranous colitis. Epidemiological changes in CDIs have been observed in the last two decades, with the emergence of highly virulent types and more numerous and severe CDI cases in the community. C. difficile interacts with the gut microbiota throughout its entire life cycle, and the C. difficile's role as colonizer or invader largely depends on alterations in the gut microbiota, which C. difficile itself can promote and maintain. The restoration of the gut microbiota to a healthy state is considered potentially effective for the prevention and treatment of CDI. Besides a fecal microbiota transplantation (FMT), many other approaches to re-establishing intestinal eubiosis are currently under investigation. This review aims to explore current data on C. difficile and gut microbiota changes in colonized individuals and infected patients with a consideration of the recent emergence of highly virulent C. difficile types, with an overview of the microbial interventions used to restore the human gut microbiota.

19.
Food Sci Biotechnol ; 33(9): 2065-2080, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39130661

RESUMEN

The human gut microbiome accompanies us from birth, and it is developed and matured by diet, lifestyle, and environmental factors. During aging, the bacterial composition evolves in reciprocal communication with the host's physiological properties. Many diseases are closely related to the gut microbiome, which means the modulation of the gut microbiome can promote the disease targeting remote organs. This review explores the intricate interaction between the gut microbiome and other organs, and their improvement from disease by prebiotics, probiotics, synbiotics, and postbiotics. Each section of the review is supported by clinical trials that substantiate the benefits of modulation the gut microbiome through dietary intervention for improving primary health outcomes across various axes with the gut. In conclusion, the review underscores the significant potential of targeting the gut microbiome for therapeutic and preventative interventions in a wide range of diseases, calling for further research to fully unlock the microbiome's capabilities in enhancing human health.

20.
Mol Nutr Food Res ; : e2400429, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39194379

RESUMEN

SCOPE: Inflammatory bowel disease (IBD) is one of the most common chronic and debilitating functional bowel disorders affecting around 11% of the population across the world. IBD is associated with 3.6 million physician visits per year, being the most common reason visiting a gastroenterologist and the second most common reason to be absent from work, sharply increasing the health care costs. METHODS AND RESULTS: Several treatments seem to be effective in IBD symptoms relief, such as probiotics, prebiotics, fecal microbiota transplantation (FMT), and dietary patterns. Probiotics (living microorganisms that can be supplemented) can protect against pathogenic bacteria due to their antimicrobial qualities. Prebiotics (nondigestible food ingredients) promote the growth of beneficial microbial strains in the gut, giving a health benefit to the host. FMT is supposed to directly change the recipient's microbial composition when a transfer of gastrointestinal microbiota from a healthy donor is carried out. And finally, dietary patterns are in the spotlight, due to the presence of certain nutrients in the gastrointestinal tract affecting gastrointestinal motility, sensitivity, barrier function, and gut microbiota. CONCLUSION: It is particularly important to know what treatment options are available and which are the most efficient in relieving IBD symptoms and improving IBD patient's quality of life.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA