18 research outputs found

    Stan odżywienia chorych po transplantacji komórek krwiotwórczych

    Get PDF
    Niedożywienie jest istotnym problemem dotykającym chorych poddanych transplantacji komórek krwiotwórczych (HCT). W okresie wczesnym po HCT, powikłania ze strony przewodu pokarmowego po postępowaniu przygotowawczym prowadzą do zmniejszonej podaży pokarmu i zaburzeń wchłaniania. Nasileniu niedożywienia sprzyja znacznie zwiększony katabolizm oraz rozwój choroby przeszczep przeciw gospodarzowi. Prowadzenie optymalnego leczenia żywieniowego pozostaje istotnym elementem opieki potransplantacyjnej. W pracy przedstawiono znaczenia regularnej oceny stanu odżywienia biorców HCT oraz omówiono dostępne metody leczenia żywieniowego. Żywienie doustne, które należy kontynuować tak długo jak jest to możliwe, można uzupełnić o preparaty wysokobiałkowe i wysokokaloryczne. Jeżeli żywienie doustne nie jest możliwe lub nie pokrywa zapotrzebowanie białkowo-kalorycznego, można rozważyć żywienie dojelitowe, jednak w praktyce klinicznej najczęściej stosowane jest żywienie pozajelitowe. W leczeniu żywieniowym chorych po HCT zastosowanie mogą znaleźć składniki immunomodulujące, w tym glutamina i kwasy tłuszczowe omega-3, oraz prebiotyki i probiotyki, które mogą korzystnie wpłynąć na skład mikrobiomu jelit

    Insights into oral microbiome and colorectal cancer – on the way of searching new perspectives

    Get PDF
    Microbiome is a keystone polymicrobial community that coexist with human body in a beneficial relationship. These microorganisms enable the human body to maintain homeostasis and take part in mechanisms of defense against infection and in the absorption of nutrients. Even though microbiome is involved in physiologic processes that are beneficial to host health, it may also cause serious detrimental issues. Additionally, it has been proven that bacteria can migrate to other human body compartments and colonize them even although significant structural differences with the area of origin exist. Such migrations have been clearly observed when the causes of genesis and progression of colorectal cancer (CRC) have been investigated. It has been demonstrated that the oral microbiome is capable of penetrating into the large intestine and cause impairments leading to dysbiosis and stimulation of cancerogenic processes. The main actors of such events seem to be oral pathogenic bacteria belonging to the red and orange complex (regarding classification of bacteria in the context of periodontal diseases), such as Porphyromonas gingivalis and Fusobacterium nucleatum respectively, which are characterized by significant amount of cancerogenic virulence factors. Further examination of oral microbiome and its impact on CRC may be crucial on early detection of this disease and would allow its use as a precise non-invasive biomarker

    Gut Microbiome Modulation and Faecal Microbiota Transplantation Following Allogenic Hematopoietic Stem Cell Transplantation

    No full text
    Nowadays, allogenic hematopoietic stem cell transplantation (allo-HSCT) is a curative therapy that is mainly recommended for hematologic malignancies. However, complications (such as graft-versus-host disease, mucositis, disease relapse, and infections) associated with the HSCT procedure contribute to the development of gut microbiota imbalance, gut-barrier disruption, and increased intestinal permeability. In the present narrative review, the crosstalk between gut microbiota products and intestinal homeostasis is discussed. Notably, gut-microbiota-related aspects have an impact on patients’ clinical outcomes and overall survival. In accordance with the most recent published data, gut microbiota is crucial for the treatment effectiveness of many diseases, not only gastrointestinal cancers but also hematologic malignancies. Therefore, it is necessary to indicate a therapeutic method allowing to modulate gut microbiota in HSCT recipients. Currently, fecal microbiota transplantation (FMT) is the most innovative method used to alter/restore gut microbiota composition, as well as modulate its activity. Despite the fact that some previous data have shown promising results, the knowledge regarding FMT in HSCT is still strongly limited, except for the treatment of Clostridium difficile infection. Additionally, administration of prebiotics, probiotics, synbiotics, and postbiotics can also modify gut microbiota; however, this strategy should be considered carefully due to the high risk of fungemia/septicemia (especially in case of fungal probiotics)

    Gut Microbiota Modulation in the Context of Immune-Related Aspects of Lactobacillus spp. and Bifidobacterium spp. in Gastrointestinal Cancers

    No full text
    Accumulating evidence has revealed the critical roles of commensal microbes in cancer progression and recently several investigators have evaluated the therapeutic effectiveness of targeting the microbiota. This gut microbiota-related approach is especially attractive in the treatment of gastrointestinal cancers. Probiotics supplementation is a microbiota-targeted strategy that appears to improve treatment efficacy; Lactobacillus spp. and Bifidobacterium spp. are among the most commonly used probiotic agents. These bacteria seem to exert immunomodulatory effects, impacting on the immune system both locally and systemically. The gut microbiota are able to affect the efficiency of immunotherapy, mainly acting as inhibitors at immune checkpoints. The effects of immunotherapy may be modulated using traditional probiotic strains and/or next generation probiotics, such as Akkermansia municiphila. It is possible that probiotics might enhance the efficiency of immunotherapy based on PD-1/PD-L1 and CTLA-4 but more data are needed to confirm this speculation. Indeed, although there is experimental evidence for the efficacy of several strains, the health-promoting effects of numerous probiotics have not been demonstrated in human patients and furthermore the potential risks of these products, particularly in oncologic patients, are rarely mentioned

    The use of probiotics in prevention and treatment of gastric and colorectal cancer

    No full text
    The composition of gut microbiota depends on many factors, such as age, life style (eating habits and the level of physical activity), pharmacological treatment (antibiotics, side effects of anti-cancer therapy) as well as surgical procedures. The gut microbiota is involved in carcinogenesis process. Furthermore, gut dysbiosis is described as qualitative and quantitative alterations in gut microbiota and it is observed in cancers. For instance, in patients with colorectal cancer the increased amount of Fusobacterium nucleatum, Bacteroides fragilis, Enterococcus faecalis, Streptococcus bovis as well as Peptostreptococcus anaerobius is noted. It was confirmed that amount of several specific bacteria, such as Lactobacillus, Escherichia-Shigella, Nitrospirae, Burkholderia fungorum and Lachnospiraceae, is increased in patients with gastric cancer. However, the major carcinogen involved in gastric carcinogenesis is Helicobacter pylori; it causes mucosa inflammation, mucosa atrophy, and as a consequence development of gastric cancer. Nowadays, there are several therapeutic methods, which may be used to alter the composition and the activity of gut microbiota. They include administration of probiotic strains, prebiotics, and synbiotics. Probiotics can be used to prevent the development of gastric and colorectal cancer, which was shown in many in vivo and in vitro studies. According to the most recent trials, probiotics reduce the incidence of diarrhoea associated with enteral nutrition. Probiotic strains may also be used as a supportive therapy in treatment of Helicobacter pylori infection. Notwithstanding, they can play a supportive role in standard eradication treatment due to reduction of adverse events of antibiotics. Probiotics decrease the incidence of infections in postooperative period, the frequency of abdominal pain, and radiation-induced diarrhoea. To sum up, probiotics may be used to prevent the development of cancer and they may significantly improve the efficiency of standard anti-cancer therapy.Mikrobiota przewodu pokarmowego jest modyfikowana przez wiele czynników, w tym styl życia, leczenie farmakologiczne oraz zabiegi chirurgiczne. Dysbioza jelitowa, czyli zaburzenia w składzie i aktywności mikrobioty, może wystąpić w przebiegu chorób nowotworowych. U pacjentów z rakiem jelita grubego obserwuje się zwiększone ilości bakterii – Fusobacterium nucleatum, Bacteroides fragilis, Enterococcus faecalis, Streptococcus bovis oraz Peptostreptococcus anaerobius. Największym karcynogenem raka żołądka jest Helicobacter pylori. Bakteria ta powoduje zapalenie błony śluzowej żołądka prowadząc do jej atrofii, a następnie do rozwinięcia nowotworu. Obecnie znanych jest kilka metod terapeutycznych modyfikujących mikrobiotę przewodu pokarmowego, w tym podaż szczepów probiotycznych, prebiotyków oraz synbiotyków. Probiotyki mogą być stosowane w profilaktyce oraz leczeniu raka żołądka i jelita grubego, co zostało potwierdzone w badaniach in vivo oraz in vitro. Według najnowszych doniesień, probiotyki są skuteczne w zmniejszeniu częstości występowania biegunki będącej skutkiem ubocznym żywienia enteralnego. Dotychczasowe badania potwierdzają także, że szczepy probiotyczne nie mogą być stosowane jako jedyny czynnik eradykacyjny Helicobacter pylori, ale stanowią terapię uzupełniającą podczas standardowego leczenia oraz redukują działania niepożądane antybiotykoterapii. Z kolei u chorych z rakiem jelita grubego probiotyki zmniejszają ryzyko rozwinięcia infekcji pooperacyjnych, bólów brzucha i biegunki związanej z radioterapią. Podsumowując, należy podkreślić, że probiotyki mają zastosowanie na etapie profilaktyki chorób nowotworowych, a także mogą znacząco poprawić wyniki standardowego leczenia przeciwnowotworowego

    Gut-Muscle Axis Exists and May Affect Skeletal Muscle Adaptation to Training

    No full text
    Excessive training may limit physiological muscle adaptation through chronic oxidative stress and inflammation. Improper diet and overtraining may also disrupt intestinal homeostasis and in consequence enhance inflammation. Altogether, these factors may lead to an imbalance in the gut ecosystem, causing dysregulation of the immune system. Therefore, it seems to be important to optimize the intestinal microbiota composition, which is able to modulate the immune system and reduce oxidative stress. Moreover, the optimal intestinal microbiota composition may have an impact on muscle protein synthesis and mitochondrial biogenesis and function, as well as muscle glycogen storage. Aproperly balanced microbiome may also reduce inflammatory markers and reactive oxygen species production, which may further attenuate macromolecules damage. Consequently, supplementation with probiotics may have some beneficial effect on aerobic and anaerobic performance. The phenomenon of gut-muscle axis should be continuously explored to function maintenance, not only in athletes

    The Modification of the Gut Microbiota via Selected Specific Diets in Patients with Crohn’s Disease

    No full text
    Gastrointestinal symptoms in Crohn’s disease (CD) are common and affect the quality of life of patients; consequently, a growing number of studies have been published on diet interventions in this group. The role of the gut microbiota in the pathogenesis and the progression of inflammatory bowel diseases (IBD), including CD, has been widely discussed. Mainly, a decreased abundance of Firmicutes, species of the Bifidobacterium genus, and the Faecalibacterium prausnitzii species as well as a reduced general diversity have been described. In this review article, we summarize available data on the influence of reduction diets on the microbiome of patients with CD. One of the most frequently used elimination diets in CD patients is the low-FODMAP (Fermentable Oligosaccharides, Disaccharides, Monosaccharides, and Polyols) diet. Although many papers show it may reduce abdominal pain, diarrhea, or bloating, it also reduces the intake of prebiotic substances, which can negatively affect the gut microbiota composition, decreasing the abundance of Bifidobacterium species and Faecalibacterium prausnitzii. Other elimination diets used by IBD patients, such as lactose-free or gluten-free diets, have also been shown to disturb the microbial diversity. On the other hand, CDED (Crohn’s disease exclusion diet) with partial enteral nutrition not only induces the remission of CD but also has a positive influence on the microbiota. The impact of diet interventions on the microbiota and, potentially, on the future course of the disease should be considered when nutritional guidelines for IBD patients are designed. Dietetic recommendations should be based not only on the regulation of the symptoms but also on the long-term development of the disease

    Gut Biofactory—Neurocompetent Metabolites within the Gastrointestinal Tract. A Scoping Review

    No full text
    The gut microbiota have gained much scientific attention recently. Apart from unravelling the taxonomic data, we should understand how the altered microbiota structure corresponds to functions of this complex ecosystem. The metabolites of intestinal microorganisms, especially bacteria, exert pleiotropic effects on the human organism and contribute to the host systemic balance. These molecules play key roles in regulating immune and metabolic processes. A subset of them affect the gut brain axis signaling and balance the mental wellbeing. Neurotransmitters, short chain fatty acids, tryptophan catabolites, bile acids and phosphatidylcholine, choline, serotonin, and L-carnitine metabolites possess high neuroactive potential. A scoping literature search in PubMed/Embase was conducted up until 20 June 2020, using three major search terms “microbiota metabolites” AND “gut brain axis” AND “mental health”. This review aimed to enhance our knowledge regarding the gut microbiota functional capacity, and support current and future attempts to create new compounds for future clinical interventions

    Fungal Gut Microbiota Dysbiosis and Its Role in Colorectal, Oral, and Pancreatic Carcinogenesis

    No full text
    The association between bacterial as well as viral gut microbiota imbalance and carcinogenesis has been intensively analysed in many studies; nevertheless, the role of fungal gut microbiota (mycobiota) in colorectal, oral, and pancreatic cancer development is relatively new and undiscovered field due to low abundance of intestinal fungi as well as lack of well-characterized reference genomes. Several specific fungi amounts are increased in colorectal cancer patients; moreover, it was observed that the disease stage is strongly related to the fungal microbiota profile; thus, it may be used as a potential diagnostic biomarker for adenomas. Candida albicans, which is the major microbe contributing to oral cancer development, may promote carcinogenesis via several mechanisms, mainly triggering inflammation. Early detection of pancreatic cancer provides the opportunity to improve survival rate, therefore, there is a need to conduct further studies regarding the role of fungal microbiota as a potential prognostic tool to diagnose this cancer at early stage. Additionally, growing attention towards the characterization of mycobiota may contribute to improve the efficiency of therapeutic methods used to alter the composition and activity of gut microbiota. The administration of Saccharomyces boulardii in oncology, mainly in immunocompromised and/or critically ill patients, is still controversial
    corecore