Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 281
Filtrar
1.
Probiotics Antimicrob Proteins ; 14(2): 224-237, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35031968

RESUMEN

Probiotics are living microorganisms that have favorable effects on human and animal health. The most usual types of microorganisms recruited as probiotics are lactic acid bacteria (LAB) and bifidobacteria. To date, numerous utilizations of probiotics have been reported. In this paper, it is suggested that probiotic bacteria can be recruited to remove and degrade different types of toxins such as mycotoxins and algal toxins that damage host tissues and the immune system causing local and systemic infections. These microorganisms can remove toxins by disrupting, changing the permeability of the plasma membrane, producing metabolites, inhibiting the protein translation, hindering the binding to GTP binding proteins to GM1 receptors, or by preventing the interaction between toxins and adhesions. Here, we intend to review the mechanisms that probiotic bacteria use to eliminate and degrade microbial toxins.


Asunto(s)
Micotoxinas , Probióticos , Animales , Bacterias/metabolismo , Bifidobacterium , Sistema Inmunológico/microbiología , Micotoxinas/metabolismo
2.
Nutrients ; 13(12)2021 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-34959752

RESUMEN

Intestinal colonization of the neonate is highly dependent on the term of pregnancy, the mode of delivery, the type of feeding [breast feeding or formula feeding]. Postnatal immune maturation is dependent on the intestinal microbiome implementation and composition and type of feeding is a key issue in the human gut development, the diversity of microbiome, and the intestinal function. It is well established that exclusive breastfeeding for 6 months or more has several benefits with respect to formula feeding. The composition of the new generation of infant formulas aims in mimicking HM by reproducing its beneficial effects on intestinal microbiome and on the gut associated immune system (GAIS). Several approaches have been developed currently for designing new infant formulas by the addition of bioactive ingredients such as human milk oligosaccharides (HMOs), probiotics, prebiotics [fructo-oligosaccharides (FOSs) and galacto-oligosaccharides (GOSs)], or by obtaining the so-called post-biotics also known as milk fermentation products. The aim of this article is to guide the practitioner in the understanding of these different types of Microbiota Influencing Formulas by listing and summarizing the main concepts and characteristics of these different models of enriched IFs with bioactive ingredients.


Asunto(s)
Ingestión de Alimentos/inmunología , Microbioma Gastrointestinal/inmunología , Sistema Inmunológico/microbiología , Fórmulas Infantiles/química , Fenómenos Fisiológicos Nutricionales del Lactante/inmunología , Femenino , Humanos , Sistema Inmunológico/crecimiento & desarrollo , Fórmulas Infantiles/microbiología , Recién Nacido , Intestinos/crecimiento & desarrollo , Intestinos/inmunología , Masculino , Leche Humana/química , Leche Humana/microbiología , Oligosacáridos/administración & dosificación , Prebióticos/administración & dosificación
3.
Front Immunol ; 12: 770436, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34970260

RESUMEN

The article presents the function of platelets in inflammation as well as in bacterial and viral infections, which are the result of their reaction with the endovascular environment, including cells of damaged vascular endothelium and cells of the immune system. This role of platelets is conditioned by biologically active substances present in their granules and in their specific structures - EV (extracellular vesicles).


Asunto(s)
Infecciones Bacterianas/inmunología , Plaquetas/inmunología , Vesículas Extracelulares/inmunología , Sistema Inmunológico/inmunología , Inflamación/inmunología , Virosis/inmunología , Infecciones Bacterianas/microbiología , Plaquetas/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Humanos , Sistema Inmunológico/microbiología , Sistema Inmunológico/virología , Inflamación/metabolismo , Mediadores de Inflamación/inmunología , Mediadores de Inflamación/metabolismo , Modelos Inmunológicos , Transducción de Señal/inmunología , Virosis/virología
4.
Int J Mol Sci ; 22(22)2021 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-34830149

RESUMEN

Fungi represent one of the most diverse and abundant eukaryotes on earth. The interplay between mold exposure and the host immune system is still not fully elucidated. Literature research focusing on up-to-date publications is providing a heterogenous picture of evidence and opinions regarding the role of mold and mycotoxins in the development of immune diseases. While the induction of allergic immune responses by molds is generally acknowledged, other direct health effects like the toxic mold syndrome are controversially discussed. However, recent observations indicate a particular importance of mold/mycotoxin exposure in individuals with pre-existing dysregulation of the immune system, due to exacerbation of underlying pathophysiology including allergic and non-allergic chronic inflammatory diseases, autoimmune disorders, and even human immunodeficiency virus (HIV) disease progression. In this review, we focus on the impact of mycotoxins regarding their impact on disease progression in pre-existing immune dysregulation. This is complemented by experimental in vivo and in vitro findings to present cellular and molecular modes of action. Furthermore, we discuss hypothetical mechanisms of action, where evidence is missing since much remains to be discovered.


Asunto(s)
Hongos/inmunología , Hipersensibilidad/inmunología , Sistema Inmunológico/inmunología , Micotoxinas/inmunología , Contaminantes Atmosféricos/análisis , Contaminantes Atmosféricos/envenenamiento , Animales , Asma/etiología , Asma/inmunología , Asma/microbiología , Exposición a Riesgos Ambientales/efectos adversos , Exposición a Riesgos Ambientales/análisis , Hongos/fisiología , Humanos , Hipersensibilidad/etiología , Hipersensibilidad/microbiología , Sistema Inmunológico/efectos de los fármacos , Sistema Inmunológico/microbiología , Micosis/etiología , Micosis/inmunología , Micosis/microbiología , Micotoxinas/envenenamiento
5.
Nutrients ; 13(11)2021 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-34836095

RESUMEN

The gut microbiota is a crucial factor in maintaining homeostasis. The presence of commensal microorganisms leads to the stimulation of the immune system and its maturation. In turn, dysbiosis with an impaired intestinal barrier leads to accelerated contact of microbiota with the host's immune cells. Microbial structural parts, i.e., pathogen-associated molecular patterns (PAMPs), such as flagellin (FLG), peptidoglycan (PGN), lipoteichoic acid (LTA), and lipopolysaccharide (LPS), induce inflammation via activation of pattern recognition receptors. Microbial metabolites can also develop chronic low-grade inflammation, which is the cause of many metabolic diseases. This article aims to systematize information on the influence of microbiota on chronic inflammation and the benefits of microbiota modification through dietary changes, prebiotics, and probiotic intake. Scientific research indicates that the modification of the microbiota in various disease states can reduce inflammation and improve the metabolic profile. However, since there is no pattern for a healthy microbiota, there is no optimal way to modify it. The methods of influencing microbiota should be adapted to the type of dysbiosis. Although there are studies on the microbiota and its effects on inflammation, this subject is still relatively unknown, and more research is needed in this area.


Asunto(s)
Disbiosis/inmunología , Microbioma Gastrointestinal/inmunología , Sistema Inmunológico/microbiología , Inflamación/microbiología , Enfermedad Crónica , Homeostasis , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Prebióticos/administración & dosificación , Probióticos/uso terapéutico , Simbiosis/inmunología
6.
Nutrients ; 13(11)2021 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-34836121

RESUMEN

Within the last two decades tremendous efforts in biomedicine have been undertaken to understand the interplay of commensal bacteria living in and on our human body with our own human physiology. It became clear that (1) a high diversity especially of the microbial communities in the gut are important to preserve health and that (2) certain bacteria via nutrition-microbe-host metabolic axes are beneficially affecting various functions of the host, including metabolic control, energy balance and immune function. While a large set of evidence indicate a special role for small chain fatty acids (SCFA) in that context, recently also metabolites of amino acids (e.g., tryptophan and arginine) moved into scientific attention. Of interest, microbiome alterations are not only important in nutrition associated diseases like obesity and diabetes, but also in many chronic inflammatory, oncological and neurological abnormalities. From a clinician's point of view, it should be mentioned, that the microbiome is not only interesting to develop novel therapies, but also as a modifiable factor to improve efficiency of modern pharmaceutics, e.g., immune-therapeutics in oncology. However, so far, most data rely on animal experiments or human association studies, whereas controlled clinical intervention studies are spare. Hence, the translation of the knowledge of the last decades into clinical routine will be the challenge of microbiome based biomedical research for the next years. This review aims to provide examples for future clinical applications in various entities and to suggest bacterial species and/or microbial effector molecules as potential targets for intervention studies.


Asunto(s)
Enfermedad Crónica , Microbiota , Investigación Biomédica Traslacional/tendencias , Animales , Metabolismo Energético , Microbioma Gastrointestinal , Humanos , Sistema Inmunológico/microbiología , Inflamación/microbiología , Fenómenos Fisiológicos de la Nutrición
7.
Nutrients ; 13(10)2021 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-34684638

RESUMEN

Chronic kidney disease (CKD) is generally progressive and irreversible, structural or functional renal impairment for 3 or more months affecting multiple metabolic pathways. Recently, the composition, dynamics, and stability of a patient's microbiota has been noted to play a significant role during disease onset or progression. Increasing urea concentration during CKD can lead to an acceleration of the process of kidney injury leading to alterations in the intestinal microbiota that can increase the production of gut-derived toxins and alter the intestinal epithelial barrier. A detailed analysis of the relationship between the role of intestinal microbiota and the development of inflammation within the symbiotic and dysbiotic intestinal microbiota showed significant changes in kidney dysfunction. Several recent studies have determined that dietary factors can significantly influence the activation of immune cells and their mediators. Moreover, dietary changes can profoundly affect the balance of gut microbiota. The aim of this review is to present the importance and factors influencing the differentiation of the human microbiota in the progression of kidney diseases, such as CKD, IgA nephropathy, idiopatic nephropathy, and diabetic kidney disease, with particular emphasis on the role of the immune system. Moreover, the effects of nutrients, bioactive compounds on the immune system in development of chronic kidney disease were reviewed.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Sistema Inmunológico/microbiología , Fenómenos Fisiológicos de la Nutrición/inmunología , Insuficiencia Renal Crónica/inmunología , Insuficiencia Renal Crónica/microbiología , Humanos , Riñón/inmunología , Riñón/microbiología
9.
Int J Mol Sci ; 22(12)2021 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-34199182

RESUMEN

The gut microbiota has been known to modulate the immune responses in chronic liver diseases. Recent evidence suggests that effects of dietary foods on health care and human diseases are related to both the immune reaction and the microbiome. The gut-microbiome and intestinal immune system play a central role in the control of bacterial translocation-induced liver disease. Dysbiosis, small intestinal bacterial overgrowth, translocation, endotoxemia, and the direct effects of metabolites are the main events in the gut-liver axis, and immune responses act on every pathways of chronic liver disease. Microbiome-derived metabolites or bacteria themselves regulate immune cell functions such as recognition or activation of receptors, the control of gene expression by epigenetic change, activation of immune cells, and the integration of cellular metabolism. Here, we reviewed recent reports about the immunologic role of gut microbiotas in liver disease, highlighting the role of diet in chronic liver disease.


Asunto(s)
Dieta , Microbioma Gastrointestinal/inmunología , Sistema Inmunológico/microbiología , Hepatopatías/inmunología , Hepatopatías/microbiología , Animales , Humanos
10.
Cells ; 10(5)2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34069602

RESUMEN

Trogocytosis is an active process, in which one cell extracts the cell fragment from another cell, leading to the transfer of cell surface molecules, together with membrane fragments. Recent reports have revealed that trogocytosis can modulate various biological responses, including adaptive and innate immune responses and homeostatic responses. Trogocytosis is evolutionally conserved from protozoan parasites to eukaryotic cells. In some cases, trogocytosis results in cell death, which is utilized as a mechanism for antibody-dependent cytotoxicity (ADCC). In other cases, trogocytosis-mediated intercellular protein transfer leads to both the acquisition of novel functions in recipient cells and the loss of cellular functions in donor cells. Trogocytosis in immune cells is typically mediated by receptor-ligand interactions, including TCR-MHC interactions and Fcγ receptor-antibody-bound molecule interactions. Additionally, trogocytosis mediates the transfer of MHC molecules to various immune and non-immune cells, which confers antigen-presenting activity on non-professional antigen-presenting cells. In this review, we summarize the recent advances in our understanding of the role of trogocytosis in immune modulation.


Asunto(s)
Comunicación Celular , Endocitosis , Sistema Inmunológico/metabolismo , Animales , Muerte Celular , Antígenos de Histocompatibilidad/metabolismo , Interacciones Huésped-Patógeno , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/microbiología , Sistema Inmunológico/patología , Ligandos , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de IgG/metabolismo , Receptores de Células Asesinas Naturales/metabolismo , Transducción de Señal
11.
Cell ; 184(15): 3884-3898.e11, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34143954

RESUMEN

Immune-microbe interactions early in life influence the risk of allergies, asthma, and other inflammatory diseases. Breastfeeding guides healthier immune-microbe relationships by providing nutrients to specialized microbes that in turn benefit the host's immune system. Such bacteria have co-evolved with humans but are now increasingly rare in modern societies. Here we show that a lack of bifidobacteria, and in particular depletion of genes required for human milk oligosaccharide (HMO) utilization from the metagenome, is associated with systemic inflammation and immune dysregulation early in life. In breastfed infants given Bifidobacterium infantis EVC001, which expresses all HMO-utilization genes, intestinal T helper 2 (Th2) and Th17 cytokines were silenced and interferon ß (IFNß) was induced. Fecal water from EVC001-supplemented infants contains abundant indolelactate and B. infantis-derived indole-3-lactic acid (ILA) upregulated immunoregulatory galectin-1 in Th2 and Th17 cells during polarization, providing a functional link between beneficial microbes and immunoregulation during the first months of life.


Asunto(s)
Bifidobacterium/fisiología , Sistema Inmunológico/crecimiento & desarrollo , Sistema Inmunológico/microbiología , Antibacterianos/farmacología , Biomarcadores/metabolismo , Lactancia Materna , Linfocitos T CD4-Positivos/inmunología , Polaridad Celular , Proliferación Celular , Citocinas/metabolismo , Heces/química , Heces/microbiología , Galectina 1/metabolismo , Microbioma Gastrointestinal , Humanos , Indoles/metabolismo , Recién Nacido , Inflamación/sangre , Inflamación/genética , Mucosa Intestinal/inmunología , Metaboloma , Leche Humana/química , Oligosacáridos/metabolismo , Células Th17/inmunología , Células Th2/inmunología , Agua
13.
Int J Mol Sci ; 22(6)2021 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-33802883

RESUMEN

The importance of the gut microbiota in human health is currently well established. It contributes to many vital functions such as development of the host immune system, digestion and metabolism, barrier against pathogens or brain-gut communication. Microbial colonization occurs during infancy in parallel with maturation of the host immune system; therefore, an adequate cross-talk between these processes is essential to generating tolerance to gut microbiota early in life, which is crucial to prevent allergic and immune-mediated diseases. Inflammatory bowel disease (IBD) is characterized by an exacerbated immune reaction against intestinal microbiota. Changes in abundance in the gut of certain microorganisms such as bacteria, fungi, viruses, and archaea have been associated with IBD. Microbes that are commonly found in high abundance in healthy gut microbiomes, such as F. prausnitzii or R. hominis, are reduced in IBD patients. E. coli, which is usually present in a healthy gut in very low concentrations, is increased in the gut of IBD patients. Microbial taxa influence the immune system, hence affecting the inflammatory status of the host. This review examines the IBD microbiome profile and presents IBD as a model of dysbiosis.


Asunto(s)
Microbioma Gastrointestinal , Sistema Inmunológico/microbiología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Animales , Disbiosis/microbiología , Humanos , Hipótesis de la Higiene , Sistema Inmunológico/crecimiento & desarrollo , Intestinos/crecimiento & desarrollo , Intestinos/microbiología , Intestinos/patología
14.
Nutrients ; 13(4)2021 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-33923663

RESUMEN

The main objective of this research was to carry out an experimental study, triple-blind, on the possible immunophysiological effects of a nutritional supplement (synbiotic, Gasteel Plus®, Heel España S.A.U.), containing a mixture of probiotic strains, such as Bifidobacterium lactis CBP-001010, Lactobacillus rhamnosus CNCM I-4036, and Bifidobacterium longum ES1, as well as the prebiotic fructooligosaccharides, on both professional athletes and sedentary people. The effects on some inflammatory/immune (IL-1ß, IL-10, and immunoglobulin A) and stress (epinephrine, norepinephrine, dopamine, serotonin, corticotropin-releasing hormone (CRH), Adrenocorticotropic hormone (ACTH), and cortisol) biomarkers were evaluated, determined by flow cytometer and ELISA. The effects on metabolic profile and physical activity, as well as on various parameters that could affect physical and mental health, were also evaluated via the use of accelerometry and validated questionnaires. The participants were professional soccer players in the Second Division B of the Spanish League and sedentary students of the same sex and age range. Both study groups were randomly divided into two groups: a control group-administered with placebo, and an experimental group-administered with the synbiotic. Each participant was evaluated at baseline, as well as after the intervention, which lasted one month. Only in the athlete group did the synbiotic intervention clearly improve objective physical activity and sleep quality, as well as perceived general health, stress, and anxiety levels. Furthermore, the synbiotic induced an immunophysiological bioregulatory effect, depending on the basal situation of each experimental group, particularly in the systemic levels of IL-1ß (increased significantly only in the sedentary group), CRH (decreased significantly only in the sedentary group), and dopamine (increased significantly only in the athlete group). There were no significant differences between groups in the levels of immunoglobulin A or in the metabolic profile as a result of the intervention. It is concluded that synbiotic nutritional supplements can improve anxiety, stress, and sleep quality, particularly in sportspeople, which appears to be linked to an improved immuno-neuroendocrine response in which IL-1ß, CRH, and dopamine are clearly involved.


Asunto(s)
Sistema Inmunológico/microbiología , Sistemas Neurosecretores/microbiología , Fútbol/fisiología , Estrés Psicológico/microbiología , Simbióticos/administración & dosificación , Acelerometría , Adulto , Ansiedad/sangre , Ansiedad/microbiología , Ansiedad/terapia , Atletas/psicología , Bifidobacterium animalis , Bifidobacterium longum , Biomarcadores/sangre , Hormona Liberadora de Corticotropina/sangre , Dopamina/sangre , Ejercicio Físico , Femenino , Humanos , Interleucina-1beta/sangre , Lacticaseibacillus rhamnosus , Masculino , Oligosacáridos/administración & dosificación , Proyectos Piloto , Probióticos/administración & dosificación , Proyectos de Investigación , Conducta Sedentaria , Sueño , Estrés Psicológico/sangre , Estrés Psicológico/terapia , Estudiantes/psicología , Encuestas y Cuestionarios , Adulto Joven
15.
Front Immunol ; 12: 644269, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33815397

RESUMEN

The first 1000 days of life, including the intrauterine period, are regarded as a fundamental stepping stone for the development of a human. Unequivocally, nutrition during this period plays a key role on the proper development of a child, both directly through the intake of essential nutrients and indirectly by affecting the composition of the gut microbiota. The gut microbiota, including bacteria, viruses, fungi, protists and other microorganisms, is a highly modifiable and adaptive system that is influenced by diet, lifestyle, medicinal products and the environment. Reversely, it affects the immune system in multiple complex ways. Many noncommunicable diseases (NCDs) associated with dysbiosis are "programmed" during childhood. Nutrition is a potent determinant of the children's microbiota composition and maturation and, therefore, a strong determinant of the NCDs' programming. In this review we explore the interplay between nutrition during the first 1000 days of life, the gut microbiota, virome and mycobiome composition and the development of NCDs.


Asunto(s)
Fenómenos Fisiológicos Nutricionales Infantiles/inmunología , Microbioma Gastrointestinal/inmunología , Sistema Inmunológico , Micobioma/inmunología , Enfermedades no Transmisibles , Viroma/inmunología , Niño , Humanos , Sistema Inmunológico/crecimiento & desarrollo , Sistema Inmunológico/microbiología
16.
Nutrients ; 13(3)2021 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-33803407

RESUMEN

Infectious diseases and infections remain a leading cause of death in low-income countries and a major risk to vulnerable groups, such as infants and the elderly. The immune system plays a crucial role in the susceptibility, persistence, and clearance of these infections. With 70-80% of immune cells being present in the gut, there is an intricate interplay between the intestinal microbiota, the intestinal epithelial layer, and the local mucosal immune system. In addition to the local mucosal immune responses in the gut, it is increasingly recognized that the gut microbiome also affects systemic immunity. Clinicians are more and more using the increased knowledge about these complex interactions between the immune system, the gut microbiome, and human pathogens. The now well-recognized impact of nutrition on the composition of the gut microbiota and the immune system elucidates the role nutrition can play in improving health. This review describes the mechanisms involved in maintaining the intricate balance between the microbiota, gut health, the local immune response, and systemic immunity, linking this to infectious diseases throughout life, and highlights the impact of nutrition in infectious disease prevention and treatment.


Asunto(s)
Enfermedades Transmisibles/inmunología , Enfermedades Transmisibles/microbiología , Microbioma Gastrointestinal/fisiología , Sistema Inmunológico/microbiología , Fenómenos Fisiológicos de la Nutrición/inmunología , Anciano , Femenino , Humanos , Inmunidad Mucosa , Lactante , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Masculino
17.
Nutrients ; 13(3)2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33804415

RESUMEN

Breastfeeding is the gold standard for feeding infants because of its long-term benefits to health and development, but most infants in the United States are not exclusively breastfed in the first six months. We enrolled 24 infants who were either exclusively breastfed or supplemented with formula by the age of one month. We collected diet information, stool samples for evaluation of microbiotas by 16S rRNA sequencing, and blood samples for assessment of immune development by flow cytometry from birth to 6 months of age. We further typed the Bifidobacterium strains in stool samples whose 16S rRNA sequencing showed the presence of Bifidobacteriaceae. Supplementation with formula during breastfeeding transiently changed the composition of the gut microbiome, but the impact dissipated by six months of age. For example, Bifidobacterium longum, a bacterial species highly correlated with human milk consumption, was found to be significantly different only at 1 month of age but not at later time points. No immunologic differences were found to be associated with supplementation, including the development of T-cell subsets, B cells, or monocytes. These data suggest that early formula supplementation, given in addition to breast milk, has minimal lasting impact on the gut microbiome or immunity.


Asunto(s)
Suplementos Dietéticos/microbiología , Microbioma Gastrointestinal/inmunología , Sistema Inmunológico/crecimiento & desarrollo , Fórmulas Infantiles/microbiología , Fenómenos Fisiológicos Nutricionales del Lactante/inmunología , Lactancia Materna/métodos , Encuestas sobre Dietas , Heces/microbiología , Femenino , Humanos , Sistema Inmunológico/microbiología , Lactante , Recién Nacido , Masculino , ARN Ribosómico 16S/aislamiento & purificación , Estados Unidos
18.
Biochem Soc Trans ; 49(2): 617-627, 2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33704415

RESUMEN

The human skin microbiota forms a key barrier against skin pathogens and is important in modulating immune responses. Recent studies identify lactobacilli as endogenous inhabitants of healthy skin, while inflammatory skin conditions are often associated with a disturbed skin microbiome. Consequently, lactobacilli-based probiotics are explored as a novel treatment of inflammatory skin conditions through their topical skin application. This review focuses on the potential beneficial role of lactobacilli (family Lactobacillaceae) in the skin habitat, where they can exert multifactorial local mechanisms of action against pathogens and inflammation. On one hand, lactobacilli have been shown to directly compete with skin pathogens through adhesion inhibition, production of antimicrobial metabolites, and by influencing pathogen metabolism. The competitive anti-pathogenic action of lactobacilli has already been described mechanistically for common different skin pathogens, such as Staphylococcus aureus, Cutibacterium acnes, and Candida albicans. On the other hand, lactobacilli also have an immunomodulatory capacity associated with a reduction in excessive skin inflammation. Their influence on the immune system is mediated by bacterial metabolites and cell wall-associated or excreted microbe-associated molecular patterns (MAMPs). In addition, lactobacilli can also enhance the skin barrier function, which is often disrupted as a result of infection or in inflammatory skin diseases. Some clinical trials have already translated these mechanistic insights into beneficial clinical outcomes, showing that topically applied lactobacilli can temporarily colonize the skin and promote skin health, but more and larger clinical trials are required to generate in vivo mechanistic insights and in-depth skin microbiome analysis.


Asunto(s)
Antibiosis/inmunología , Candida albicans/inmunología , Inflamación/inmunología , Lactobacillus/inmunología , Piel/inmunología , Staphylococcus aureus/inmunología , Antibiosis/fisiología , Adhesión Bacteriana/inmunología , Bacteriocinas/inmunología , Bacteriocinas/metabolismo , Candida albicans/fisiología , Humanos , Sistema Inmunológico/inmunología , Sistema Inmunológico/microbiología , Inflamación/microbiología , Lactobacillus/metabolismo , Lactobacillus/fisiología , Piel/microbiología , Piel/patología , Staphylococcus aureus/fisiología
19.
Curr Top Dev Biol ; 141: 399-427, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33602495

RESUMEN

Animals have evolved within the framework of the microbes and are constantly exposed to diverse microbiota. This dominance of the microbial world is forcing all fields of biology to question some of their most basic premises, with developmental biology being no exception. While animals under laboratory conditions can develop and live without microbes, they are far from normal, and would not survive under natural conditions, where their fitness would be strongly compromised. Since much of the undescribed biodiversity on Earth is microbial, any consideration of animal development in the absence of the recognition of microbes will be incomplete. Here, we show that animal development may never have been autonomous, rather it requires transient or persistent interactions with the microbial world. We propose that to formulate a comprehensive understanding of embryogenesis and post-embryonic development, we must recognize that symbiotic microbes provide important developmental signals and contribute in significant ways to phenotype production. This offers limitless opportunities for the field of developmental biology to expand.


Asunto(s)
Evolución Biológica , Eucariontes/citología , Gastrulación/fisiología , Simbiosis , Animales , Bacterias/citología , Biopelículas , Femenino , Microbioma Gastrointestinal , Sistema Inmunológico/microbiología , Masculino , Rumiantes/microbiología
20.
FEBS Lett ; 595(9): 1322-1327, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33570779

RESUMEN

The gut microbiota and the immune system have co-evolved to interact and cooperate in many ways. A recent study characterizing the immunomodulatory effects of over 60 different human-derived gut microbes across phyla showed that bacteria-induced immunomodulations are not dictated by the bacterial phylogeny. Yet, it remains unclear whether strains from the same species induce the same immunomodulatory effects on the host. We analyzed the strain-level data from this recent study and found that strains from the same species can induce distinct and sometimes even opposing immunophenotypes. Hence, we suggest that the immunomodulatory capabilities of gut bacteria can be strain-specific.


Asunto(s)
Microbioma Gastrointestinal/genética , Tracto Gastrointestinal/microbiología , Inmunomodulación/genética , Filogenia , Bacterias/genética , Bacterias/inmunología , Microbioma Gastrointestinal/inmunología , Humanos , Sistema Inmunológico/metabolismo , Sistema Inmunológico/microbiología , ARN Ribosómico 16S/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...