Gut Microbiota and Antipsychotics Induced Metabolic Alteration

Author(s):  
Dong-Yu Kan ◽  
Su-Juan Li ◽  
Chen-Chen Liu ◽  
Ren-Rong Wu

Schizophrenia is a chronic and severe mental disorder with antipsychotics as primary medications, but the antipsychotic-induced metabolic side effects may contribute to the elevated risk of overall morbidity and mortality in patients with psych-iatric diseases. With the development in sequencing technology and bioinformatics, dysbiosis has been shown to contribute to body weight gain and metabolic dysfunction. However, the role of gut microbiota in the antipsychotic-induced metabolic alteration remains unknown. In this paper, we reviewed the recent studies of the gut microbiota with psychiatric disorders and antipsychotic-induced metabolic dysfunction. Patients with neuropsychiatric disorders may have a different composi-tion of gut microbiota compared with healthy controls. In addition, it seems that the use of antipsychotics is concurrently associated with both altered composition of gut microbiota and metabolic disturbance. Further study is needed to address the role of gut microbiota in the development of neuropsychiatric disorders and antipsychotic-induced metabolic disturbance, to develop novel therapeutics for both neuropsychiatric disorders and metabolic dysfunction.

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Cai Tan ◽  
Zhihua Zheng ◽  
Xiaogang Wan ◽  
Jiaqing Cao ◽  
Ran Wei ◽  
...  

AbstractThe change in gut microbiota is an important mechanism of the amelioration of type 2 diabetes mellitus (T2DM) after bariatric surgery. Here, we observe that the modified jejunoileal bypass effectively decreases body weight gain, fasting blood glucose, and lipids level in serum; additionally, islet β-cell function, glucose tolerance, and insulin resistance were markedly ameliorated. The hypoglycemic effect and the improvement in islet β-cell function depend on the changes in gut microbiota structure. modified jejunoileal bypass increases the abundance of gut Escherichia coli and Ruminococcus gnavus and the levels of serum glycine, histidine, and glutamine in T2DM rats; and decreases the abundance of Prevotella copri and the levels of serum branched chain amino acids, which are significantly related to the improvement of islet β-cell function in T2DM rats. Our results suggest that amino acid metabolism may contribute to the islet β-cell function in T2DM rats after modified jejunoileal bypass and that improving gut microbiota composition is a potential therapeutic strategy for T2DM.


2020 ◽  
Vol 8 (2) ◽  
pp. e000964 ◽  
Author(s):  
Bogang Wu ◽  
Huai-Chin Chiang ◽  
Xiujie Sun ◽  
Bin Yuan ◽  
Payal Mitra ◽  
...  

The programmed death-ligand 1 (PD-L1)-dependent immune checkpoint attenuates host immunity and maintains self-tolerance. Imbalance between protective immunity and immunopathology due to altered PD-L1 signaling can lead to autoimmunity or tumor immunosuppression. The role of the PD-L1-dependent checkpoint in non-immune system is less reported. We previously found that white adipocytes highly express PD-L1. Here we show that adipocyte-specific PD-L1 knockout mice exhibit enhanced host anti-tumor immunity against mammary tumors and melanoma with low or no tumor PD-L1. However, adipocyte PD-L1 ablation in tumor-free mice also exacerbates diet-induced body weight gain, pro-inflammatory macrophage infiltration into adipose tissue, and insulin resistance. Low PD-L1 mRNA levels in human adipose tissue correlate with high body mass index and presence of type 2 diabetes. Therefore, our mouse genetic approach unequivocally demonstrates a cell-autonomous function of adipocyte PD-L1 in promoting tumor growth and inhibiting antitumor immunity. In addition, our work uncovers a previously unrecognized role of adipocyte PD-L1 in mitigating obesity-related inflammation and metabolic dysfunction.


2018 ◽  
Vol 2018 ◽  
pp. 1-10 ◽  
Author(s):  
Yiqiang Xie ◽  
Man Xiao ◽  
Yali Ni ◽  
Shangfei Jiang ◽  
Guizhu Feng ◽  
...  

Recently, the role of gut microbiota in the development of obesity and type 2 diabetes mellitus (T2DM) has been highlighted. We performed an 8-week administration protocol on T2DM (C57BL/6J db-/db-) mice and fecal samples were collected. Comparisons of fecal bacterial communities were performed between db-/db- mice and normal mice (DB/DB) and between the db-/db mice treated and untreated with AOE using next-generation sequencing technology. Our results showed that the db-/db-AOE group had improved glycemic control and renal function compared with the db-/db-H2O group. Compared with the db-/db-H2O group, AOE administration resulted in significantly increased ratio of Bacteroidetes-to-Firmicutes in db-/db- mice. In addition, the abundance ofAkkermansiawas significantly increased, whileHelicobacterwas significantly suppressed in the db-/db-AOE group compared with the db-/db-H2O group. Our data suggest that AOE treatment decreased blood glucose levels and significantly reduced damage of renal pathology in the T2DM mice by modulating gut microbiota composition.


Life ◽  
2021 ◽  
Vol 11 (8) ◽  
pp. 760
Author(s):  
Donatella Marazziti ◽  
Beatrice Buccianelli ◽  
Stefania Palermo ◽  
Elisabetta Parra ◽  
Alessandro Arone ◽  
...  

The functioning of the central nervous system (CNS) is the result of the constant integration of bidirectional messages between the brain and peripheral organs, together with their connections with the environment. Despite the anatomical separation, gut microbiota, i.e., the microorganisms colonising the gastrointestinal tract, is highly related to the CNS through the so-called “gut–brain axis”. The aim of this paper was to review and comment on the current literature on the role of the intestinal microbiota and the gut–brain axis in some common neuropsychiatric conditions. The recent literature indicates that the gut microbiota may affect brain functions through endocrine and metabolic pathways, antibody production and the enteric network while supporting its possible role in the onset and maintenance of several neuropsychiatric disorders, neurodevelopment and neurodegenerative disorders. Alterations in the gut microbiota composition were observed in mood disorders and autism spectrum disorders and, apparently to a lesser extent, even in obsessive-compulsive disorder (OCD) and related conditions, as well as in schizophrenia. Therefore, gut microbiota might represent an interesting field of research for a better understanding of the pathophysiology of common neuropsychiatric disorders and possibly as a target for the development of innovative treatments that some authors have already labelled “psychobiotics”.


2021 ◽  
Vol 11 (9) ◽  
pp. 304-309
Author(s):  
Sara Moqbil ◽  
Sylwiusz Niedobylski ◽  
Katarzyna Laszczak ◽  
Konrad Warchoł ◽  
Eryk Mikos

INTRODUCTION AND PURPOSE Recent studies have shown that changes in the microbiome, probiotic and antibiotic supplementation, can significantly modulate various forms of neuropsychiatric disorders - such as depression, anxiety and stress-related disorders. There is growing body of evidence pointing to a bidirectional correlation along the brain-gut microbiota line. This axis is connected through endocrine, immune and neuronal pathways. The nerves that make up the enteric nervous system transmit modifications occurring in the gastrointestinal tract and through the vagus nerve to the central nervous system (1). The main purpose of this review is to update recent information on the correlation between the gut microbiota and mental health.  STATE OF KNOWLEDGE Interactions between the gastrointestinal system and brain function have become an important field of psychiatric research in recent years. Probiotics are thought to be a potentially valuable player in the treatment of many neuropsychiatric disorders. However, the role of specific gut microbiota species in the development of these disorders remains unclear. CONCLUSIONS Increasing knowledge of the correlation between gut microbiota and mental health may improve the quality of treatment for patients with neuropsychiatric conditions. Further research on larger groups is needed to assess whether probiotics can modify altered psychological well-being and be integrated into current, conventional treatments.


Nutrients ◽  
2021 ◽  
Vol 13 (3) ◽  
pp. 713
Author(s):  
Jeanne Alard ◽  
Benoit Cudennec ◽  
Denise Boutillier ◽  
Véronique Peucelle ◽  
Amandine Descat ◽  
...  

Since alterations of the gut microbiota have been shown to play a major role in obesity, probiotics have attracted attention. Our aim was to identify probiotic candidates for the management of obesity using a combination of in vitro and in vivo approaches. We evaluated in vitro the ability of 23 strains to limit lipid accumulation in adipocytes and to enhance the secretion of satiety-promoting gut peptide in enteroendocrine cells. Following the in vitro screening, selected strains were further investigated in vivo, single, or as mixtures, using a murine model of diet-induced obesity. Strain Bifidobacterium longum PI10 administrated alone and the mixture of B. animalis subsp. lactis LA804 and Lactobacillus gasseri LA806 limited body weight gain and reduced obesity-associated metabolic dysfunction and inflammation. These protective effects were associated with changes in the hypothalamic gene expression of leptin and leptin receptor as well as with changes in the composition of gut microbiota and the profile of bile acids. This study provides crucial clues to identify new potential probiotics as effective therapeutic approaches in the management of obesity, while also providing some insights into their mechanisms of action.


2020 ◽  
Vol 22 (2) ◽  
pp. 36-44
Author(s):  
Magdalena Nalewajska ◽  
Marcin Lisak ◽  
Małgorzata Marchelek – Myśliwiec ◽  
Violetta Dziedziejko ◽  
Andrzej Pawlik

2021 ◽  
Author(s):  
Devindra Shakappa ◽  
Aruna Talari ◽  
Rakesh Naik

Abstract The legume prebiotics, especially galacto-oligosaccharides, are the center of health since they play a crucial role in maintaining gut health. The aim of the present study was to understand the role of prebiotic galacto-oligosaccharides of pigeon pea (Cajanus cajan L) on gut microbiota, obesity and type 2 diabetes mellitus in Wistar National Institute of Nutrition NIN rats. Proximate composition and oligosaccharide content of the pigeon pea was estimated and for animal experiment, four-week-old weaning male Wistar National Institute of Nutrition NIN rats (N = 36) were divided equally into three groups (n = 12) control, raffinose, and pigeon pea prebiotics groups. All the groups were fed with the respective diet for 18 weeks. After 18 weeks 5 ml of blood was drawn from the retro-orbital plexus and the rats were sacrificed by (CO2) inhalation. Among the studied groups, the pigeon pea prebiotic supplementation significantly (p˂0.05) lowered body weight gain (284.7 ± 36.4 grams) and resulted in lower body mass index (3.79 ± 0.18kg / m2). The fat percent was significantly lower in pigeon pea prebiotic fed group (7.8 ± 2.7) than raffinose (11.6 ± 3.1) and control (13.8 ± 2) groups. Pigeon pea prebiotic feeding significantly (< 0.05) lowered blood glucose levels; it also improved the Homeostasis model assessment of insulin resistance (HOMA IR) index in pigeon pea prebiotic group (32.08 ± 5.37). Pigeon pea prebiotic feeding significantly (p˂0.05) enhanced the fold increase of lactobacillus and bifidobacterial count when compared to raffinose fed group. The present study explores the role of pigeon pea prebiotics on gut microbiota and the regulation of some of the features of metabolic syndrome such as obesity and diabetes.


2020 ◽  
Vol 19 (7) ◽  
pp. 509-526
Author(s):  
Qin Huang ◽  
Fang Yu ◽  
Di Liao ◽  
Jian Xia

: Recent studies implicate microbiota-brain communication as an essential factor for physiology and pathophysiology in brain function and neurodevelopment. One of the pivotal mechanisms about gut to brain communication is through the regulation and interaction of gut microbiota on the host immune system. In this review, we will discuss the role of microbiota-immune systeminteractions in human neurological disorders. The characteristic features in the development of neurological diseases include gut dysbiosis, the disturbed intestinal/Blood-Brain Barrier (BBB) permeability, the activated inflammatory response, and the changed microbial metabolites. Neurological disorders contribute to gut dysbiosis and some relevant metabolites in a top-down way. In turn, the activated immune system induced by the change of gut microbiota may deteriorate the development of neurological diseases through the disturbed gut/BBB barrier in a down-top way. Understanding the characterization and identification of microbiome-immune- brain signaling pathways will help us to yield novel therapeutic strategies by targeting the gut microbiome in neurological disease.


Sign in / Sign up

Export Citation Format

Share Document