Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 110
Filter
2.
Article in English | LILACS, VETINDEX | ID: biblio-1518148

ABSTRACT

Studies on the interactions between the intestinal microbiome and its host have strengthened in the last decade. However, publications on this topic in dogs still need to be made available, reinforcing the need for new studies and literary data for consultation. Given this, this review aims to describe the intestinal microbiome and its interactions with the canine host, which can contribute to both health and morbid conditions in these animals. The definition of microbiome encompasses the collective genome of all microorganisms that live in a defined habitat (intestine). It is known that the dog's intestinal microbiota is varied, composed of bacteria, archaea, viruses, fungi, and protozoa. Under normal conditions, there is commensalism between some of these microorganisms and the host, which promotes critical physiological relationships and interactions that contribute to homeostasis and the consequent health of the animal. With this in mind, it is expected that the disturbances associated with the microbiome will result in imbalances in this commensal relationship and thus precipitate the development of diseases and aggravation of other diseases, thus characterizing intestinal dysbiosis.(AU)


Os estudos sobre as interações entre o microbioma intestinal e o seu hospedeiro ganharam força na última década. Entretanto, as publicações acerca de tal temática em cães ainda são escassas, o que reforça a necessidade de novos estudos e dados literários para consultas. Frente a isso, o objetivo da presente revisão é descrever sobre o microbioma intestinal e suas interações e principais efeitos no cão, os quais podem contribuir tanto para a higidez quanto para quadros mórbidos desses animais. A definição de microbioma engloba o genoma coletivo de todos os microrganismos que vivem em habitat definido (intestino). É sabido que a microbiota intestinal do cão é muito variada, sendo composta por bactérias, arqueas, vírus, fungos e protozoários. Em condições normais, há o comensalismo entre alguns desses microrganismos e o hospedeiro, o que promove importantes relações e interações fisiológicas que contribuem sobremaneira para a homeostasia e consequente saúde do animal. Ciente disso, é de se esperar que os distúrbios associados ao microbioma resultarão em desequilíbrios nessa relação comensal e, assim, precipitar o desenvolvimento de doenças e/ou agravamento de outras moléstias, caracterizando, assim, a disbiose intestinal.(AU)


Subject(s)
Animals , Dogs/microbiology , Gastrointestinal Microbiome , Dysbiosis
3.
International Journal of Oral Science ; (4): 4-4, 2023.
Article in English | WPRIM | ID: wpr-971592

ABSTRACT

Periodontitis is the most widespread oral disease and is closely related to the oral microbiota. The oral microbiota is adversely affected by some pharmacologic treatments. Systemic antibiotics are widely used for infectious diseases but can lead to gut dysbiosis, causing negative effects on the human body. Whether systemic antibiotic-induced gut dysbiosis can affect the oral microbiota or even periodontitis has not yet been addressed. In this research, mice were exposed to drinking water containing a cocktail of four antibiotics to explore how systemic antibiotics affect microbiota pathogenicity and oral bone loss. The results demonstrated, for the first time, that gut dysbiosis caused by long-term use of antibiotics can disturb the oral microbiota and aggravate periodontitis. Moreover, the expression of cytokines related to Th17 was increased while transcription factors and cytokines related to Treg were decreased in the periodontal tissue. Fecal microbiota transplantation with normal mice feces restored the gut microbiota and barrier, decreased the pathogenicity of the oral microbiota, reversed the Th17/Treg imbalance in periodontal tissue, and alleviated alveolar bone loss. This study highlights the potential adverse effects of long-term systemic antibiotics-induced gut dysbiosis on the oral microbiota and periodontitis. A Th17/Treg imbalance might be related to this relationship. Importantly, these results reveal that the periodontal condition of patients should be assessed regularly when using systemic antibiotics in clinical practice.


Subject(s)
Humans , Mice , Animals , Dysbiosis , Anti-Bacterial Agents/pharmacology , Virulence , Microbiota , Periodontitis/chemically induced , Cytokines
4.
Braz. j. biol ; 83: e242818, 2023. tab, graf
Article in English | LILACS, VETINDEX | ID: biblio-1285628

ABSTRACT

Abstract The study was aimed to assess impact of high fat diet (HFD) and synthetic human gut microbiota (GM) combined with HFD and chow diet (CD) in inducing type-2 diabetes (T2D) using mice model. To our knowledge, this is the first study using selected human GM transplantation via culture based method coupled dietary modulation in mice for in vivo establishment of inflammation leading to T2D and gut dysbiosis. Twenty bacteria (T2D1-T2D20) from stool samples of confirmed T2D subjects were found to be morphologically different and subjected to purification on different media both aerobically and anerobically, which revealed seven bacteria more common among 20 isolates on the basis of biochemical characterization. On the basis of 16S rRNA gene sequencing, these seven isolates were identified as Bacteroides stercoris (MT152636), Lactobacillus acidophilus (MT152637), Lactobacillus salivarius (MT152638), Ruminococcus bromii (MT152639), Klebsiella aerogenes (MT152640), Bacteroides fragilis (MT152909), Clostridium botulinum (MT152910). The seven isolates were subsequently used as synthetic gut microbiome (GM) for their role in inducing T2D in mice. Inbred strains of albino mice were divided into four groups and were fed with CD, HFD, GM+HFD and GM+CD. Mice receiving HFD and GM+modified diet (CD/HFD) showed highly significant (P<0.05) increase in weight and blood glucose concentration as well as elevated level of inflammatory cytokines (TNF-α, IL-6, and MCP-1) compared to mice receiving CD only. The 16S rRNA gene sequencing of 11 fecal bacteria obtained from three randomly selected animals from each group revealed gut dysbiosis in animals receiving GM. Bacterial strains including Bacteroides gallinarum (MT152630), Ruminococcus bromii (MT152631), Lactobacillus acidophilus (MT152632), Parabacteroides gordonii (MT152633), Prevotella copri (MT152634) and Lactobacillus gasseri (MT152635) were isolated from mice treated with GM+modified diet (HFD/CD) compared to strains Akkermansia muciniphila (MT152625), Bacteriodes sp. (MT152626), Bacteroides faecis (MT152627), Bacteroides vulgatus (MT152628), Lactobacillus plantarum (MT152629) which were isolated from mice receiving CD/HFD. In conclusion, these findings suggest that constitution of GM and diet plays significant role in inflammation leading to onset or/and possibly progression of T2D. .


Resumo O estudo teve como objetivo avaliar o impacto da dieta rica em gordura (HFD) e da microbiota intestinal humana sintética (GM) combinada com HFD e dieta alimentar (CD) na indução de diabetes tipo 2 (T2D) usando modelo de camundongos. Para nosso conhecimento, este é o primeiro estudo usando transplante de GM humano selecionado através do método baseado em cultura acoplada à modulação dietética em camundongos para o estabelecimento in vivo de inflamação que leva a T2D e disbiose intestinal. Vinte bactérias (T2D1-T2D20) de amostras de fezes de indivíduos T2D confirmados verificaram ser morfologicamente diferentes e foram submetidas à purificação em meios diferentes aerobicamente e anaerobicamente, o que revelou sete bactérias mais comuns entre 20 isolados com base na caracterização bioquímica. Com base no sequenciamento do gene 16S rRNA, esses sete isolados foram identificados como Bacteroides stercoris (MT152636), Lactobacillus acidophilus (MT152637), Lactobacillus salivarius (MT152638), Ruminococcus bromii (MT152639), Klebsiella aerogenides (MT152640), Bacteroides fragilis (MT152909), Clostridium botulinum (MT152910). Esses sete isolados foram, posteriormente, usados ​​como microbioma intestinal sintético (GM) por seu papel na indução de T2D em camundongos. Linhagens consanguíneas de camundongos albinos foram divididas em quatro grupos e foram alimentadas com CD, HFD, GM + HFD e GM + CD. Camundongos que receberam a dieta modificada com HFD e GM + (CD / HFD) mostraram um aumento altamente significativo (P < 0,05) no peso e na concentração de glicose no sangue, bem como um nível elevado de citocinas inflamatórias (TNF-α, IL-6 e MCP-1) em comparação com os ratos que receberam apenas CD. O sequenciamento do gene 16S rRNA de 11 bactérias fecais obtidas de três animais selecionados aleatoriamente de cada grupo revelou disbiose intestinal em animais que receberam GM. Cepas bacterianas, incluindo Bacteroides gallinarum (MT152630), Ruminococcus bromii (MT152631), Lactobacillus acidophilus (MT152632), Parabacteroides gordonii (MT152633), Prevotella copri (MT152634) e Lactobacillus Gasseri (MT152635D), foram tratadas com dieta modificada / CD) em comparação com as linhagens Akkermansia muciniphila (MT152625), Bacteriodes sp. (MT152626), Bacteroides faecis (MT152627), Bacteroides vulgatus (MT152628), Lactobacillus plantarum (MT152629), que foram isoladas de camundongos recebendo CD / HFD. Em conclusão, esses resultados sugerem que a constituição de GM e dieta desempenham papel significativo na inflamação levando ao início ou/e possivelmente à progressão de T2D.


Subject(s)
Humans , Animals , Rabbits , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Bacteroides , RNA, Ribosomal, 16S/genetics , Prevotella , Bacteroidetes , Ruminococcus , Diet, High-Fat/adverse effects , Dysbiosis , Inflammation , Mice, Inbred C57BL
5.
Braz. j. biol ; 83: 1-14, 2023. ilus, tab, graf
Article in English | LILACS, VETINDEX | ID: biblio-1468843

ABSTRACT

The study was aimed to assess impact of high fat diet (HFD) and synthetic human gut microbiota (GM) combined with HFD and chow diet (CD) in inducing type-2 diabetes (T2D) using mice model. To our knowledge, this is the first study using selected human GM transplantation via culture based method coupled dietary modulation in mice for in vivo establishment of inflammation leading to T2D and gut dysbiosis. Twenty bacteria (T2D1-T2D20) from stool samples of confirmed T2D subjects were found to be morphologically different and subjected to purification on different media both aerobically and anerobically, which revealed seven bacteria more common among 20 isolates on the basis of biochemical characterization. On the basis of 16S rRNA gene sequencing, these seven isolates were identified as Bacteroides stercoris (MT152636), Lactobacillus acidophilus (MT152637), Lactobacillus salivarius (MT152638), Ruminococcus bromii (MT152639), Klebsiella aerogenes (MT152640), Bacteroides fragilis (MT152909), Clostridium botulinum (MT152910). The seven isolates were subsequently used as synthetic gut microbiome (GM) for their role in inducing T2D in mice. Inbred strains of albino mice were divided into four groups and were fed with CD, HFD, GM+HFD and GM+CD. Mice receiving HFD and GM+modified diet (CD/HFD) showed highly significant (P<0.05) increase in weight and blood glucose concentration as well as elevated level of inflammatory cytokines (TNF-α, IL-6, and MCP-1) compared to mice receiving CD only. The 16S rRNA gene sequencing of 11 fecal bacteria obtained from three randomly selected animals from each group revealed gut dysbiosis in animals receiving GM. Bacterial strains including Bacteroides gallinarum (MT152630), Ruminococcus bromii (MT152631), Lactobacillus acidophilus (MT152632), Parabacteroides gordonii (MT152633), Prevotella copri (MT152634) and Lactobacillus gasseri (MT152635) were isolated from mice [...].


O estudo teve como objetivo avaliar o impacto da dieta rica em gordura (HFD) e da microbiota intestinal humana sintética (GM) combinada com HFD e dieta alimentar (CD) na indução de diabetes tipo 2 (T2D) usando modelo de camundongos. Para nosso conhecimento, este é o primeiro estudo usando transplante de GM humano selecionado através do método baseado em cultura acoplada à modulação dietética em camundongos para o estabelecimento in vivo de inflamação que leva a T2D e disbiose intestinal. Vinte bactérias (T2D1-T2D20) de amostras de fezes de indivíduos T2D confirmados verificaram ser morfologicamente diferentes e foram submetidas à purificação em meios diferentes aerobicamente e anaerobicamente, o que revelou sete bactérias mais comuns entre 20 isolados com base na caracterização bioquímica. Com base no sequenciamento do gene 16S rRNA, esses sete isolados foram identificados como Bacteroides stercoris (MT152636), Lactobacillus acidophilus (MT152637), Lactobacillus salivarius (MT152638), Ruminococcus bromii (MT152639), Klebsiella aerogenides (MT152640), Bacteroides fragilis (MT152909), Clostridium botulinum (MT152910). Esses sete isolados foram, posteriormente, usados como microbioma intestinal sintético (GM) por seu papel na indução de T2D em camundongos. Linhagens consanguíneas de camundongos albinos foram divididas em quatro grupos e foram alimentadas com CD, HFD, GM + HFD e GM + CD. Camundongos que receberam a dieta modificada com HFD e GM + (CD / HFD) mostraram um aumento altamente significativo (P < 0,05) no peso e na concentração de glicose no sangue, bem como um nível elevado de citocinas inflamatórias (TNF-α, IL-6 e MCP-1) em comparação com os ratos que receberam apenas CD. O sequenciamento do gene 16S rRNA de 11 bactérias fecais obtidas de três animais selecionados aleatoriamente de cada grupo revelou disbiose intestinal em animais que receberam GM. Cepas bacterianas, incluindo Bacteroides gallinarum (MT152630), Ruminococcus [...].


Subject(s)
Humans , Adult , Mice , /etiology , /prevention & control , /veterinary , Dysbiosis/veterinary , Dietary Fats/adverse effects , Gastrointestinal Microbiome
6.
Journal of Zhejiang University. Science. B ; (12): 430-441, 2023.
Article in English | WPRIM | ID: wpr-982383

ABSTRACT

Early weaned piglets suffer from oxidative stress and enteral infection, which usually results in gut microbial dysbiosis, serve diarrhea, and even death. Rice bran oil (RBO), a polyphenol-enriched by-product of rice processing, has been shown to have antioxidant and anti-inflammatory properties both in vivo and in vitro. Here, we ascertained the proper RBO supplementation level, and subsequently determined its effects on lipopolysaccharide (LPS)-induced intestinal dysfunction in weaned piglets. A total of 168 piglets were randomly allocated into four groups of seven replicates (42 piglets each group, (21±1) d of age, body weight (7.60±0.04) kg, and half males and half females) and were given basal diet (Ctrl) or basal diet supplemented with 0.01% (mass fraction) RBO (RBO1), 0.02% RBO (RBO2), or 0.03% RBO (RBO3) for 21 d. Then, seven piglets from the Ctrl and the RBO were treated with LPS (100 μg/kg body weight (BW)) as LPS group and RBO+LPS group, respectively. Meanwhile, seven piglets from the Ctrl were treated with the saline vehicle (Ctrl group). Four hours later, all treated piglets were sacrificed for taking samples of plasma, jejunum tissues, and feces. The results showed that 0.02% was the optimal dose of dietary RBO supplementation based on diarrhea, average daily gain, and average daily feed intake indices in early weaning piglets. Furthermore, RBO protected piglets against LPS-induced jejunal epithelium damage, which was indicated by the increases in villus height, villus height/crypt depth ratio, and Claudin-1 levels, as well as a decreased level of jejunal epithelium apoptosis. RBO also improved the antioxidant ability of LPS-challenged piglets, which was indicated by the elevated concentrations of catalase and superoxide dismutase, and increased total antioxidant capacity, as well as the decreased concentrations of diamine oxidase and malondialdehyde in plasma. Meanwhile, RBO improved the immune function of LPS-challenged weaned piglets, which was indicated by elevated immunoglobulin A (IgA), IgM, β‍‍-defensin-1, and lysozyme levels in the plasma. In addition, RBO supplementation improved the LPS challenge-induced dysbiosis of gut microbiota. Particularly, the indices of antioxidant capacity, intestinal damage, and immunity were significantly associated with the RBO-regulated gut microbiota. These findings suggested that 0.02% RBO is a suitable dose to protect against LPS-induced intestinal damage, oxidative stress, and jejunal microbiota dysbiosis in early weaned piglets.


Subject(s)
Male , Female , Animals , Swine , Lipopolysaccharides/toxicity , Antioxidants/pharmacology , Rice Bran Oil , Dysbiosis , Dietary Supplements , Diarrhea/veterinary , Weaning , Body Weight
7.
Hepatología ; 4(1): 75-89, 2023. fig
Article in Spanish | LILACS, COLNAL | ID: biblio-1415978

ABSTRACT

La interrupción de la simbiosis que existe entre el cuerpo humano y su microbioma puede resultar en una disbiosis, un desequilibrio en la interacción huésped-microbiota, que puede asociarse al desarrollo de diversas enfermedades como el síndrome de intestino irritable, hígado graso no alco-hólico, enfermedad hepática alcohólica y cirrosis, entre otras. En ciertas condiciones patológicas y por múltiples factores de riesgo, la capacidad de autorregulación del intestino se puede alterar, contribuyendo al incremento de la permeabilidad con inflamación intestinal crónica. El diagnóstico y el tratamiento, así como la relación entre la permeabilidad intestinal, la disbiosis y las patologías gastrointestinales y hepatobiliares, todavía no tienen estudios clínicos validados o con el soporte científico adecuado, por lo que se realiza una revisión de la literatura con la finalidad de aportar conceptos que puedan orientar con respecto a la importancia del estudio del microbioma humano en estas enfermedades.


Disruption of the symbiosis that exists between the human body and its microbiome can result in dys-biosis, an imbalance in the host-microbiota interaction, which may be associated with the develop-ment of various diseases such as irritable bowel syndrome, non-alcoholic fatty liver disease, alcoholic liver disease and cirrhosis, among others. In certain pathological conditions and due to multiple risk factors, the self-regulating capacity of the intestine may be lost, contributing to increased permeability with chronic intestinal inflammation. Its diagnosis and treatment as well as the relationship between intestinal permeability, dysbiosis and gastrointestinal and hepatobiliary pathologies have not been validated in clinical studies or have adequate scientific support, so a review of the literature is carried out in order to provide concepts that can guide with respect to the importance of the study of the human microbiome in these diseases


Subject(s)
Humans , Permeability , Dysbiosis , Microbiota , Gastrointestinal Microbiome , Risk Factors , Irritable Bowel Syndrome , Fatty Liver , Non-alcoholic Fatty Liver Disease , Gastrointestinal Diseases , Liver Diseases, Alcoholic
8.
J. Health Biol. Sci. (Online) ; 10(1): 1-9, 01/jan./2022. ilus, tab
Article in Portuguese | LILACS | ID: biblio-1411337

ABSTRACT

Objetivos: evidenciar as relações e a existência do agravamento do Transtorno do Espectro Autista devido à disbiose intestinal. Métodos: revisão integrativa realizada segundo a pergunta norteadora: Existe comprovação científica entre a relação do TEA e disbiose intestinal que favoreça a melhora na prática clínica e indicações de possíveis respostas? Buscou-se por artigos publicados entre janeiro de 2016 e janeiro de 2021, nas bases de dados: PubMed, SciELO, LILACS, GOOGLE ACADÊMICO. Foram utilizados os descritores (DeCS): "Transtorno do Espectro Autista"; "Microbiota gastrointestinal"; "Disbiose", associados pelo operador booleano "E". Foram incluidos artigos de revisões bibliográficas, completos, originais, limitados aos idiomas inglês e português brasileiro, publicados nos últimos cinco anos, e que, após leitura do resumo, estivessem dentro do escopo da revisão. Resultados: Foram identificados 52 manuscritos e, após aplicação dos critérios de inclusão e exclusão, foram considerados 11 artigos que evidenciam o agravamento do TEA por fatores intrínsecos à microbiota intestinal. Conclusão: existe importante influência causal do eixo bidirecional cérebro-intestino-microbiota na etiologia e exacerbaçao das manifestações clínicas do Transtorno do Espectro Autista devido à disbiose intestinal e aos fatores gastrointestinais de origem idiopática.


Objectives: to highlight the relationships and the existence of Autistic Spectrum Disorder aggravation due to intestinal dysbiosis. Methods: integrative review conducted according to the guiding question: Is there scientific evidence of the relationship between ASD and intestinal dysbiosis that favors improvement in clinical practice and indications of possible answers? We searched for articles published between January 2016 and January 2021 in databases: PubMed, SciELO, LILACS, and GOOGLE ACADEMIC. The following descriptors (DeCS) were used: "Autistic Spectrum Disorder"; "Gastrointestinal microbiota"; "Dysbiosis", associated with the Boolean operator "AND". We included literature review articles, complete, original, limited to English and Brazilian Portuguese languages, published in the last five years, and which, after reading the abstract, were within the scope of the review. Results: 52 manuscripts were identified, and after applying the inclusion and exclusion criteria, 11 articles were considered that show the worsening of ASD due to factors intrinsic to the intestinal microbiota. Conclusion: there is an important causal influence of the bidirectional brain-gut-microbiota axis in the etiology and exacerbation of clinical manifestations of Autism Spectrum Disorder due to intestinal dysbiosis and gastrointestinal factors of idiopathic origin.


Subject(s)
Autism Spectrum Disorder , Alkalies , Dysbiosis , Microbiota , Gastrointestinal Microbiome , LILACS , Literature
9.
Rev. cuba. med ; 61(3)sept. 2022.
Article in Spanish | LILACS, CUMED | ID: biblio-1441681

ABSTRACT

Introducción: La microbiota describe a un grupo de microorganismos en una región o período de tiempo que incluye: bacterias, arqueas, protistas, hongos y virus. Objetivos: Explicar la función de la microbiota intestinal en la salud humana. Métodos: Se realizó una búsqueda en diferentes de bases de datos como NHI, Ebsco y PubMed en idioma español e inglés, se revisó un total de 17 artículos de los cuales el mayor por ciento es de menos de 5 años. Resultados: Las microbiota intestinal en su mayoría se compone de Gram negativa, con una pared celular rica en lipopolisacáridos (LPS) que potencia a la inmunidad innata por interacción de receptor Toll-like (TLR) ligando, desencadena la producción de citoquinas proinflamatorias, entre otros. Conclusiones: La microbiota intestinal funciona como un señalizador antiinflamatorio y regulador de la permeabilidad epitelial intestinal(AU)


Introduction: Microbiota describes a group of microorganisms in a region or period of time that includes bacteria, archaea, protists, fungi, and viruses. Objectives: To explain the role of the intestinal microbiota in human health. Methods: A search was carried out in different databases such as NHI, Ebsco and PubMed in Spanish and English, a total of 17 articles were reviewed, most of them are less than 5 years. Results: Intestinal microbiota is mostly composed of Gram negative, with a cell wall rich in lipopolysaccharides (LPS) that enhances innate immunity by Toll-like receptor (TLR) ligand interaction, triggers the production of proinflammatory cytokines, among others. Conclusions: Intestinal microbiota functions as an anti-inflammatory signaling agent and regulator of intestinal epithelial permeability(AU)


Subject(s)
Humans , Dysbiosis/microbiology , Gastrointestinal Microbiome
10.
Rev. ADM ; 79(4): 218-223, jul.-ago. 2022. tab
Article in Spanish | LILACS | ID: biblio-1396089

ABSTRACT

Objetivo: actualizar la información sobre la disbiosis bacteriana oral y su efecto en enfermedades bucales. Material y métodos: se realizó una revisión bibliográfica detallada, donde la búsqueda de artículos comenzó desde el 2014 con trabajos de investigación relacionados con el tema. Se aplicaron palabras clave para facilitar y delimitar el tema. En los resultados obtenidos se observa información específica de disbiosis bacteriana y los problemas y enfermedades que causan en la cavidad bucal. Conclusión: la cavidad oral es un ecosistema muy complejo e interactivo donde se desarrollan variedades de hábitats que establecen relaciones entre los microorganismos en los distintos medios bucales. Por lo general, el cuerpo humano vive en simbiosis con dichas bacterias, esta relación hospedador-huésped es producto de años de evolución y convivencia para poder tolerar a dichas especies y por medio de años de investigación, determinar a los agentes patógenos y a los simbióticos, lo que permitirá en un futuro tener enfoques terapéuticos y científicos, para así solucionar, mejorar y evitar problemas relacionados con la salud (AU)


Objective: this review aimed to update the information on oral bacterial dysbiosis and its effect on oral diseases. Material and methods: a detailed literature review was performed, where the search for articles began in 2014 with research papers related to the topic. Keywords were applied to facilitate and delimit the topic. The results obtained show specific information on bacterial dysbiosis and the problems and diseases they cause in the oral cavity. Conclusion: the oral cavity is a very complex and interactive ecosystem where a variety of habitats develop and establish relationships between microorganisms in different oral environments. Generally, the human body lives in symbiosis with these bacteria, this host-guest relationship is the product of years of evolution and coexistence to be able to tolerate these species and through years of research to determine the pathogens and symbiotics, which will allow in the future to have therapeutic and scientific approaches, to solve, improve and avoid health-related problems (AU)


Subject(s)
Humans , Bacterial Infections/complications , Dysbiosis/etiology , Mouth Diseases/microbiology , Gram-Positive Rods/pathogenicity , Gram-Negative Aerobic Rods and Cocci/pathogenicity , Dental Plaque/microbiology , Host Microbial Interactions , Mouth/microbiology
11.
Arch. latinoam. nutr ; 72(2): 100-108, jun. 2022. ilus
Article in Spanish | LILACS, LIVECS | ID: biblio-1381416

ABSTRACT

Las investigaciones realizadas durante el último siglo relacionadas con la descripción de la Microbiota Intestinal (MI) sugieren una relación concreta entre su composición y la salud del huésped. Su desregulación denominada disbiosis intestinal ha sido asociada a distintos tipos de enfermedades gastrointestinales, metabólicas, oncológicas e incluso psiquiátricas. Destacan numerosos reportes que han informado la condición de disbiosis en la obesidad, tanto en modelos animales como humanos de distintos grupos etarios y regiones del mundo. A su vez, la composición del microbioma también ha logrado asociarse a las diferentes comorbilidades de la obesidad, postulando que la MI posee influencia en la disfunción del tejido adiposo (TA), entendiendo que corresponde al principal modulador de la patogénesis de la obesidad. Sin embargo, aún no es posible establecer una explicación mecanicista plausible. Actualmente, la utilización de tecnologías multiómicas, junto con la evaluación de variables fisiológicas, nos podrían proporcionar una mejor comprensión a la incógnita planteada. Frente a esto, el presente trabajo tiene como objetivo revisar los últimos avances en la comprensión de la influencia de la microbiota intestinal en el TA y su contribución a los mecanismos relacionados con la patogénesis de la obesidad. Entre los principales mecanismos identificados, la evidencia reporta nexos fisiológicos entre la composición de la MI y la modulación de inflamación, permeabilidad intestinal y adipogénesis. Las vías implicadas derivan de la influencia de la disbiosis intestinal en el accionar de ácidos grasos de cadena corta, claudinas, macrófagos, oligosacáridos, entre otros. Los mecanismos implicados, principalmente estudiados en modelos animales, deberían ser considerados para su evaluación en próximos estudios longitudinales y experimentales en humanos con el fin de obtener una mayor comprensión sobre la implicancia de cada mecanismo en la patogenia global de la obesidad(AU)


The investigations carried out during the last century related to the description of the Gut Microbiota (GM) suggest a concrete relationship between its composition and the health of the host. Its deregulation called intestinal dysbiosis has been associated with different types of gastrointestinal, metabolic, oncological and even psychiatric diseases. Numerous reports that have described the condition of dysbiosis in obesity stand out, both in animal and human models of different age groups and regions of the world. In turn, the composition of the microbiome has also been associated with the different comorbidities of obesity, postulating that MI has an influence on adipose tissue (AT) dysfunction, understanding that it corresponds to the main modulator of the pathogenesis of obesity. However, it is not yet possible to establish a plausible mechanistic explanation. Currently, the use of multi-omics technologies, together with the evaluation of physiological variables, could provide us with a better understanding of the question raised. In view of this, this review aims to review the latest advances in understanding the influence of the intestinal microbiota on AT and its contribution to the mechanisms related to the pathogenesis of obesity. Among the main mechanisms identified, the evidence reports physiological links between the composition of GM and the modulation of inflammation, intestinal permeability and adipogenesis. The pathways involved derive from the influence of intestinal dysbiosis on the action of short-chain fatty acids, claudins, macrophages, oligosaccharides, among others. The mechanisms involved, mainly studied in animal models, should be considered for evaluation in future longitudinal and experimental studies in humans in order to obtain a better understanding of the implication of each mechanism in the global pathogenesis of obesity(AU)


Subject(s)
Adipose Tissue , Gastrointestinal Microbiome , Obesity/pathology , Energy Metabolism , Adipogenesis , Dysbiosis , Gastrointestinal Diseases
12.
Rev. cuba. med ; 61(1)mar. 2022.
Article in Spanish | LILACS, CUMED | ID: biblio-1408981

ABSTRACT

Introducción: La disbiosis conocida como la alteración de la relación simbiótica entre la microbiota intestinal y el huésped están implicados en la patogenia de la enfermedad cardiovascular aterosclerótica. Objetivo: Realizar una revisión documental sobre los mecanismos fisiopatológicos que relacionan los metabolitos bioactivos generados por la disbiosis intestinal con el desarrollo y progresión de la enfermedad cardiovascular aterosclerótica. Métodos: Se utilizó el motor de búsqueda Google Académico y se consultaron artículos de libre acceso en las bases de datos Pubmed, SciELO, Lilacs, Cumed y Hinari desde septiembre 2020 hasta el mes de marzo 2021. Las palabras clave utilizadas para esta revisión fueron:microbioma, microbiota intestinal, disbiosis, aterosclerosis, enfermedad cardiovascular y sus equivalentes en inglés, según el descriptor de Ciencias de la Salud (DeCS). Se consideraron artículos originales, de revisión, revisiones sistemáticas y metaanálisis posteriores al año 2015. Se revisaron un total de 73 artículos. Desarrollo: Las relaciones fisiopatológicas entre la disbiosis intestinal y las enfermedades cardiovasculares son complejas, ya que se influyen mutuamente a través de los sus toxinas endógenas (metabolitos bioactivos), el sistema circulatorio, las respuestas inmunitarias y los cambios metabólicos. Las investigaciones futuras deberían centrarse en dilucidar los actores moleculares subyacentes e identificar si las vías que interconectan la disbiosis intestinal con la ECA son causales, correlacionales o consecuentes. Conclusiones: La evidencia acumulada sostiene que la disbiosis de la microbiota intestinal está involucrada en la síntesis de metabolitos proaterogénicos los cuales modulan los mecanismos implicados en la fisiopatología de la ECA(AU)


Introduction: Dysbiosis is known as the alteration of the symbiotic relationship between the intestinal microbiota and the host is involved in the pathogenesis of atherosclerotic cardiovascular disease. Objective: To carry out a documentary review on the pathophysiological mechanisms that relate the bioactive metabolites generated by intestinal dysbiosis with the development and progression of atherosclerotic cardiovascular disease. Methods: The Google Scholar search engine was used and free access articles were consulted in Pubmed, SciELO, Lilacs, Cumed and Hinari databases from September 2020 to March 2021. The keywords used for this review were microbiome, gut microbiota, dysbiosis, atherosclerosis, cardiovascular disease and their English equivalents, according to the Health Sciences (DeCS) descriptor. Original articles, review articles, systematic reviews and meta-analyses after 2015 were considered. A total of 73 articles were reviewed. Findings: The pathophysiological relationships between intestinal dysbiosis and cardiovascular diseases are complex, since they influence each other through their endogenous toxins (bioactive metabolites), the circulatory system, immune responses and metabolic changes. Future research should focus on elucidating the underlying molecular players and on identifying whether the pathways that interconnect gut dysbiosis with ACE are causal, correlational, or consequential. Conclusions: The accumulated evidence supports that the dysbiosis of the intestinal microbiota is involved in the synthesis of proatherogenic metabolites which modulate the mechanisms involved in the pathophysiology of ACE(AU)


Subject(s)
Humans , Male , Female , Cardiovascular Diseases/epidemiology , Metabolic Syndrome/epidemiology , Atherosclerosis/epidemiology , Dysbiosis , Gastrointestinal Microbiome/physiology
13.
Alerta (San Salvador) ; 5(1): 43-49, ene. 28, 2022.
Article in Spanish | BISSAL, LILACS | ID: biblio-1354457

ABSTRACT

La incidencia de enfermedades alérgicas en la infancia va en aumento, y se ha convertido en una de las principales consultas. Una posible causa es la disbiosis del microbioma intestinal, relacionada con estados inflamatorios aumentados. Debido a la necesidad de mejorar la calidad de vida, y el impacto en lo económico y en lo educativo, surgen los probióticos como tratamiento adyuvante, por lo que se pretende determinar la asociación del uso de Bifidobacterium en menores de 5 años con la modulación de la respuesta inmune en enfermedades alérgicas. El microbioma intestinal inicia su desarrollo y maduración desde la gestación, continúa en el nacimiento y termina hasta los 3 años, influenciado por factores maternos, neonatales y ambientales. La disbiosis intestinal generada por estos factores reduce la proporción de bifidobacterias, lo cual se relaciona con estados proinflamatorios. En consecuencia, estudios del uso de Bifidobacterium en niños con enfermedades alérgicas ha evidenciado mejora de síntomas y calidad de vida. Los probióticos favorecen un microbioma intestinal saludable, asociado a un estado antiinflamatorio, debido a la regulación en el balance celular Th1/Th2/T reguladoras y células asesinas naturales. Esta modulación en la respuesta inmune permite mejor control de síntomas, calidad de vida y menor incidencia de enfermedades alérgicas en la infancia


The incidence of allergic diseases in childhood is increasing, and has become one of the main queries. One possible cause is dysbiosis of the gut microbiome, related to increased inflammatory states. Due to the need to improve the quality of life, and the economic and educational impact, probiotics emerge as adjuvant treatment, so it is intended to determine the association of the use of Bifidobacterium in children under 5 years with the modulation of the immune response in allergic diseases. The intestinal microbiome begins its development and maturation from gestation, continues at birth and ends up to 3 years, influenced by maternal, neonatal and environmental factors. The intestinal dysbiosis generated by these factors reduces the proportion of bifidobacteria, which is related to proinflammatory states. Consequently, studies of the use of Bifidobacterium in children with allergic diseases have shown improvement in symptoms and quality of life. Probiotics favor a healthy intestinal microbiome, associated with an anti-inflammatory state, due to the regulation of the regulatory Th1/Th2/T cell balance and natural killer cells. This modulation in the immune response allows better control of symptoms, quality of life and lower incidence of allergic diseases in childhood


Subject(s)
Bifidobacterium , Disease , Probiotics , Dysbiosis , Gastrointestinal Microbiome , Child , Immunity
14.
Biol. Res ; 55: 23-23, 2022. ilus, graf
Article in English | LILACS | ID: biblio-1383924

ABSTRACT

BACKGROUND: Human Gut Microbiota (HGM) is composed of more than one thousand species, playing an important role in the health status of individuals. Dysbiosis (an HGM imbalance) is augmented as chronic kidney disease (CKD) progresses, as loss of kidney function accelerates. Increased antibiotic use in CKD subjects and consumption of nephrotoxic heavy metals and metalloids such as lead, cadmium, arsenic, and mercury in tap water increases the dysbiosis state. Studies in people with stage 3 CKD are complex to carry out, mainly because patients are self-reliant who rarely consult a specialist. The current work focused on this type of patient. RESULTS: Lead and arsenic-resistant bacteria were obtained from self-reliant (that stands on its own) stage 3 CKD subjects. Pathogen-related Firmicutes and Proteobacteria genus bacteria were observed. Resistance and potentiation of antibiotic effects in the presence of metal(loid)s in vitro were found. Furthermore, the presence of the following genes markers for antibiotic and metal(loid) resistance were identified by qPCR: oxa10, qnrB1, mphB, ermB, mefE1, arr2, sulll, tetA, floR, strB, dhfr1, acrB, cadA2k, cadA3k, arsC, pbrA. We observed a decrease in the number of metal resistance markers. CONCLUSIONS: The presence of cadA and arsC genetic markers of antibiotics and metal(loid)s resistance were detected in samples from stage 3 CKD subjects. Lower gene amplification in advanced stages of CKD were also observed, possibly associated with a decrease in resident HGM during kidney disease progression.


Subject(s)
Humans , Arsenic , Metals, Heavy , Renal Insufficiency, Chronic , Gastrointestinal Microbiome , Bacteria/genetics , Drug Resistance, Microbial , Dysbiosis/microbiology , Anti-Bacterial Agents/pharmacology
15.
Chinese Journal of Stomatology ; (12): 523-528, 2022.
Article in Chinese | WPRIM | ID: wpr-935896

ABSTRACT

Emerging evidence suggests that, along with dietary, genetic and environmental factors, gut microbiota plays a role in the progress of colorectal cancer. Dysbiosis of oral flora in patients with periodontitis affects the composition of microbial community in the gut, impairs gut barrier function, and induces a proinflammatory microenvironment, all of which contribute to the progression of colorectal cancer. In view of the influences by microbiota dysbiosis, this article reviews the role of periodontitis in affecting the occurrence and development of colorectal cancer.


Subject(s)
Humans , Colorectal Neoplasms , Dysbiosis/complications , Gastrointestinal Microbiome , Microbiota , Periodontitis/complications , Tumor Microenvironment
16.
Actual. osteol ; 18(1): 40-52, 2022. ilus, tab
Article in Spanish | LILACS, UNISALUD, BINACIS | ID: biblio-1396075

ABSTRACT

El "microbioma" no solo está constituido por los microbios, sino por todos los componen-tes que viven en el mismo hábitat conforman-do un nicho ecológico. Es decir, está conformado por los microorganismos (bacterias, hongos, protozoos, etc.), todo el espectro de moléculas producidas por ellos tales como sus componentes estructurales (ácidos nucleicos, proteínas, lípidos y glúcidos), meta-bolitos, toxinas, etc., y las moléculas producidas por el huésped. El microbioma intestinal (MI) ha emergido como un factor que tiene un gran efecto sobre la cantidad, calidad y fuerza del hueso. Las investigaciones revelan que la homeostasis ósea está ligada al micro-bioma saludable, mientras que la disbiosis (alteración en la biodiversidad microbiana) puede exacerbar la actividad osteoclástica y promover la osteoporosis. Los mecanismos potenciales involucrados en la interacción del microbioma intestinal y el hueso son la influencia del metabolismo del huésped, el mantenimiento de la integridad intestinal y regulación de la absorción de nutrientes, la regulación del eje intestino-sistema inmune y la modulación del sistema endocrino. Es decir que hay múltiples vías por las cuales el MI influye sobre el hueso, pero estos y otros mecanismos deben profundizarse más aún. También es necesario que se identifiquen y caractericen mejor los microorganismos que están asociados a las enfermedades óseas. El conocimiento de estos aspectos podría ser útil para el desarrollo de herramientas terapéuticas basadas en el MI que puedan mejorar la eficacia de los distintos tratamientos existentes. (AU)


The microbiome is not only constituted by microbes, but by all the components that live in the same habitat forming an ecological niche. It is conformed by the microorganisms ( bacteria, fungi, protozoa, etc), the entire spectrum of molecules produced by them (nucleic acids, proteins, lipid and carbohydrates, metabolites, toxins, etc) and the molecules produced by the host. The intestinal microbiome (IM) has emerged as a factor with great effects on the quantity, quality and strength of bone. The investigations reveal that bone homeostasis is linked to the healthy microbiome, while the dysbiosis (alteration in the microbial biodiversity) can exacerbate the osteoclastic activity and promote osteoporosis. The potential mechanisms involved in the interaction between IM and bone are the influence of the host metabolism, the maintenance of the intestinal integrity and regulation of the nutrient absorption, the regulation of the intestine/ immune system axis and the modulation of the endocrine system. That is, there are multiple ways through which IM influences on bone, but these and other mechanisms need to be further studied. It is also necessary to identify and characterize the microorganisms associated with the bone diseases. Knowledge of these aspects could be useful to develop therapeutical tools based on the IM that could improve the efficacy of the current treatments. (AU)


Subject(s)
Humans , Osteoblasts/immunology , Osteoclasts/immunology , Bone and Bones/immunology , Dysbiosis/complications , Gastrointestinal Microbiome/immunology , Osteoblasts/metabolism , Osteoclasts/metabolism , Bone and Bones/metabolism , Intestines/immunology , Intestines/microbiology
17.
Chinese Medical Journal ; (24): 634-638, 2022.
Article in English | WPRIM | ID: wpr-927551

ABSTRACT

Inflammatory bowel disease (IBD) is a non-specific inflammatory disease of the gastrointestinal (GI) tract that is generally accepted to be closely related to intestinal dysbiosis in the host. GI infections contribute a key role in the pathogenesis of IBD; however, although the results of recent clinical studies have revealed an inverse correlation between Helicobacter pylori (H. pylori) infection and IBD, the exact mechanism underlying the development of IBD remains unclear. H. pylori, as a star microorganism, has been a focus for decades, and recent preclinical and real-world studies have demonstrated that H. pylori not only affects the changes in the gastric microbiota and microenvironment but also influences the intestinal microbiota, indicating a potential correlation with IBD. Detailed analysis revealed that H. pylori infection increased the diversity of the intestinal microbiota, reduced the abundance of Bacteroidetes, augmented the abundance of Firmicutes, and produced short-chain fatty acid-producing bacteria such as Akkermansia. All these factors may decrease vulnerability to IBD. Further studies investigating the H. pylori-intestinal microbiota metabolite axis should be performed to understand the mechanism underlying the development of IBD.


Subject(s)
Humans , Chronic Disease , Dysbiosis/microbiology , Gastrointestinal Microbiome , Helicobacter Infections , Helicobacter pylori , Inflammatory Bowel Diseases/microbiology , Microbiota
18.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 120-132, 2022.
Article in English | WPRIM | ID: wpr-929242

ABSTRACT

Wantong Jingu Tablet (WJT), a mixture of traditional Chinese medicine, was reported to relieve the symptoms of rheumatoid arthritis (RA), but its pharmacological mechanism was not completely understood. The aim of this study was to investigate the therapeutic mechanisms of WJT for RA in vivo. The effects of WJT on joint pathology, as well as the levels of Bax, Bcl-2, caspase-3, cleaved-caspase-3, ERK1/2, pERK1/2, TNF-α, IL-1β, and IL-6 were measured using collagen-induced arthritis (CIA) rats. The intestinal flora composition and the metabolites alteration were analyzed by 16S rDNA sequencing and metabolomics method, respectively. We found that WJT ameliorated the severity of the CIA rats which might be mediated by inducing apoptosis, inactivating the MEK/ERK signals and reducing the production of pro-inflammatory cytokines. WJT, in part, relieved the gut microbiota dysbiosis, especially bacterial phylum Bacteroidetes, Tenericutes and Deferribacteres, as well as bacterial genus Vibrio, Macrococcus and Vagococcus. 3'-N-debenzoyl-2'-deoxytaxol, tubulysin B, and magnoline were significantly associated with the specific genera. We identified serotonin, glutathione disulfide, N-acetylneuraminic acid, naphthalene and thromboxane B2 as targeted molecules via metabolomics. Our findings contributed to the understanding of RA pathogenesis, and WJT played essential roles in gut microbiota health and metabolite modulation in the CIA rats.


Subject(s)
Animals , Rats , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Dysbiosis , Metabolomics , Tablets
19.
Chinese Journal of Natural Medicines (English Ed.) ; (6): 443-457, 2022.
Article in English | WPRIM | ID: wpr-939910

ABSTRACT

Antibiotic exposure-induced dysbiosis of the intestinal flora increases the risk of developing allergic rhinitis. Hence, regulating the balance of intestinal flora may be useful for preventing and treating allergic rhinitis. However, the underlying mechanism is unclear. Dendrobium nobile (Shihu) exhibits anti-inflammatory and immune activities. Hence, in this study, we investigated the mechanism via which Shihu may improve allergic rhinitis. Mouse models of allergic rhinitis with intestinal flora dysbiosis (Model-D, antibiotics induce intestinal flora dysbiosis with ovalbumin-induced allergy) and normal intestinal flora with allergic rhinitis (Model-N, ovalbumin-induced allergy) were established. The effect of Shihu on intestinal flora and inflammation caused during allergic rhinitis were analyzed. Allergic symptoms, infiltration of hematoxylin and eosin in the lungs and nose, and the release of various factors [interleukin (IL)-2, IL-4, IFN-γ, IL-6, IL-10, and IL-17] in the lungs were evaluated. The results indicate that intestinal flora dysbiosis exacerbated lung and nose inflammation in allergic rhinitis. However, treatment with the Shihu extract effectively reversed these symptoms. Besides, the Shihu extract inhibited the PI3K/AKT/mTOR pathway and increased the level of Forkhead box protein in the lungs. Additionally, the Shihu extract reversed intestinal flora dysbiosis at the phylum and genus levels and improved regulator T cell differentiation. Furthermore, in the Model-D group, the Shihu extract inhibited the decrease in the diversity and abundance of the intestinal flora. Screening was performed to determine which intestinal flora was positively correlated with Treg differentiation using Spearman's correlation analysis. In conclusion, we showed that Shihu extract restored the balance in intestinal flora and ameliorated inflammation in the lungs of allergic rhinitis mice and predicted a therapeutic new approach using Traditional Chinese Medicine to improve allergic rhinitis.


Subject(s)
Animals , Mice , Cytokines/metabolism , Dendrobium , Disease Models, Animal , Drugs, Chinese Herbal/pharmacology , Dysbiosis/drug therapy , Gastrointestinal Microbiome , Inflammation/drug therapy , Mice, Inbred BALB C , Ovalbumin , Phosphatidylinositol 3-Kinases , Pneumonia , Rhinitis, Allergic/metabolism
20.
International Journal of Oral Science ; (4): 32-32, 2022.
Article in English | WPRIM | ID: wpr-939850

ABSTRACT

The aim of this study was to identify whether periodontitis induces gut microbiota dysbiosis via invasion by salivary microbes. First, faecal and salivary samples were collected from periodontally healthy participants (PH group, n = 16) and patients with severe periodontitis (SP group, n = 21) and analysed by 16S ribosomal RNA sequencing. Significant differences were observed in both the faecal and salivary microbiota between the PH and SP groups. Notably, more saliva-sourced microbes were observed in the faecal samples of the SP group. Then, the remaining salivary microbes were transplanted into C57BL6/J mice (the C-PH group and the C-SP group), and it was found that the composition of the gut microbiota of the C-SP group was significantly different from that of the C-PH group, with Porphyromonadaceae and Fusobacterium being significantly enriched in the C-SP group. In the colon, the C-SP group showed significantly reduced crypt depth and zonula occludens-1 expression. The mRNA expression levels of pro-inflammatory cytokines, chemokines and tight junction proteins were significantly higher in the C-SP group. To further investigate whether salivary bacteria could persist in the intestine, the salivary microbiota was stained with carboxyfluorescein diacetate succinimidyl ester and transplanted into mice. We found that salivary microbes from both the PH group and the SP group could persist in the gut for at least 24 h. Thus, our data demonstrate that periodontitis may induce gut microbiota dysbiosis through the influx of salivary microbes.


Subject(s)
Animals , Humans , Mice , Dysbiosis , Gastrointestinal Microbiome , Mice, Inbred C57BL , Microbiota , Periodontitis , RNA, Ribosomal, 16S/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL