scholarly journals Could Candida Overgrowth Be Involved in the Pathophysiology of Autism?

2022 ◽  
Vol 11 (2) ◽  
pp. 442
Author(s):  
Anna Herman ◽  
Andrzej Przemysław Herman

The purpose of this review is to summarize the current acquiredknowledge of Candida overgrowth in the intestine as a possible etiology of autism spectrum disorder (ASD). The influence of Candida sp. on the immune system, brain, and behavior of children with ASD isdescribed. The benefits of interventions such as a carbohydrates-exclusion diet, probiotic supplementation, antifungal agents, fecal microbiota transplantation (FMT), and microbiota transfer therapy (MTT) will be also discussed. Our literature query showed that the results of most studies do not fully support the hypothesis that Candida overgrowth is correlated with gastrointestinal (GI) problems and contributes to autism behavioral symptoms occurrence. On the one hand, it was reported that the modulation of microbiota composition in the gut may decrease Candida overgrowth, help reduce GI problems and autism symptoms. On the other hand, studies on humans suggesting the beneficial effects of a sugar-free diet, probiotic supplementation, FMT and MTT treatment in ASD are limited and inconclusive. Due to the increasing prevalence of ASD, studies on the etiology of this disorder are extremely needed and valuable. However, to elucidate the possible involvement of Candida in the pathophysiology of ASD, more reliable and well-designed research is certainly required.

Author(s):  
Ning Li ◽  
Hongyan Chen ◽  
Yi Cheng ◽  
Fenghua Xu ◽  
Guangcong Ruan ◽  
...  

Autism spectrum disorder (ASD) is a severe brain development disorder that is characterized by deficits in social communication and restricted, repetitive and stereotyped behaviors. Accumulating evidence has suggested that gut microbiota disorders play important roles in gastrointestinal symptoms and neurodevelopmental dysfunction in ASD patients. Manipulation of the gut microbiota by fecal microbiota transplantation (FMT) was recently shown to be a promising therapy for the treatment of various diseases. Here, we performed a clinical trial to evaluate the effect of FMT on gastrointestinal (GI) and ASD symptoms and gut microbiota alterations in children with ASD. We found that there was a large difference in baseline characteristics of behavior, GI symptoms, and gut microbiota between children with ASD and typically developing (TD) control children. FMT could improve GI symptoms and ASD symptoms without inducing any severe complications. Similarly, FMT significantly changed the serum levels of neurotransmitters. We further observed that FMT could promote the colonization of donor microbes and shift the bacterial community of children with ASD toward that of TD controls. The abundance of Eubacterium coprostanoligenes pre-FMT was positively correlated with high GSRS scores, whereas a decrease in Eubacterium coprostanoligenes abundance induced by FMT was associated with the FMT response. Our data suggest that FMT might be a promising therapeutic strategy to improve the GI and behavioral symptoms of patients with ASD, possibly due to its ability to alter gut microbiota and highlight a specific microbiota intervention that targets Eubacterium coprostanoligenes that can enhance the FMT response. This trial was registered at the Chinese Clinical Trial Registry (www.chictr.org.cn) (trial registration number ChiCTR1800014745).


Microbiome ◽  
2021 ◽  
Vol 9 (1) ◽  
Author(s):  
Patricia Bermudez-Martin ◽  
Jérôme A. J. Becker ◽  
Nicolas Caramello ◽  
Sebastian P. Fernandez ◽  
Renan Costa-Campos ◽  
...  

Abstract Background Autism spectrum disorders (ASD) are associated with dysregulation of the microbiota-gut-brain axis, changes in microbiota composition as well as in the fecal, serum, and urine levels of microbial metabolites. Yet a causal relationship between dysregulation of the microbiota-gut-brain axis and ASD remains to be demonstrated. Here, we hypothesized that the microbial metabolite p-Cresol, which is more abundant in ASD patients compared to neurotypical individuals, could induce ASD-like behavior in mice. Results Mice exposed to p-Cresol for 4 weeks in drinking water presented social behavior deficits, stereotypies, and perseverative behaviors, but no changes in anxiety, locomotion, or cognition. Abnormal social behavior induced by p-Cresol was associated with decreased activity of central dopamine neurons involved in the social reward circuit. Further, p-Cresol induced changes in microbiota composition and social behavior deficits could be transferred from p-Cresol-treated mice to control mice by fecal microbiota transplantation (FMT). We also showed that mice transplanted with the microbiota of p-Cresol-treated mice exhibited increased fecal p-Cresol excretion, compared to mice transplanted with the microbiota of control mice. In addition, we identified possible p-Cresol bacterial producers. Lastly, the microbiota of control mice rescued social interactions, dopamine neurons excitability, and fecal p-Cresol levels when transplanted to p-Cresol-treated mice. Conclusions The microbial metabolite p-Cresol induces selectively ASD core behavioral symptoms in mice. Social behavior deficits induced by p-Cresol are dependant on changes in microbiota composition. Our study paves the way for therapeutic interventions targeting the microbiota and p-Cresol production to treat patients with ASD.


Author(s):  
Paulina Żebrowska ◽  
Izabela Łaczmańska ◽  
Łukasz Łaczmański

Research on the use of fecal microbiota transplantation (FMT) in the treatment of disorders related to digestive system ailments in children with autism spectrum disorders (ASDs) is a new attempt in a therapeutic approach. There are very little scientific evidences available on this emerging alternative method. However, it appears to be interesting not only because of its primary outcome, relieving the gastrointestinal (GI) symptoms, but also secondary therapeutic effect of alleviating autistic behavioral symptoms. FMT seems to be also promising method in the treatment of another group of pediatric patients, children with inflammatory bowel disease (IBD). The aim of this study is to discuss the potential use of FMT and modified protocols (MTT, microbiota transfer therapy) in the treatment of GI disorders in ASD children supported by reports on another disease, IBD concerning pediatric patients. Due to the few reports of the use of FMT in the treatment of children, these two patients groups were selected, although suffering from distant health conditions: neurodevelopmental disorder and gastrointestinal tract diseases, because of the the fact that they seem related in aspects of the presence of GI symptoms, disturbed intestinal microbiota, unexplained etiology of the condition and age range of patients. Although the outcomes for all are promising, this type of therapy is still an under-researched topic, studies in the group of pediatric patients are sparse, also there is a high risk of transmission of infectious and noninfectious elements during the procedure and no long-term effects on global health are known. For those reasons all obtained results should be taken with a great caution. However, in the context of future therapeutic directions for GI observed in neurodevelopmental disorders and neurodegenerative diseases, the topic seems worthy of attention.


Author(s):  
C. Sue Carter ◽  
Suma Jacob

The effects of oxytocin and vasopressin on the brain and behavior can be sexually dimorphic, especially during the course of development (Bales, Kim, et al., 2004; Bales, Pfeifer, et al., 2004; Bales, Plotsky, et al., 2007; Bielsky et al., 2005a; Carter, 2003; Thompson et al., 2006; Yamamoto et al., 2005; Yamamoto et al., 2004). Given the sexual discrepancy observed in autism spectrum disorders (ASDs), these two neuropeptides, oxytocin (OT) and arginine vasopressin (AVP), have received attention for their potential role in ASDs (Green and Hollander, 2010; Insel et al., 1999; Leckman & Herman, 2002; Welch et al., 2005; Winslow, 2005; Young et al., 2002). Changes in either OT or AVP and their receptors could be capable of influencing symptom domains or behaviors associated with ASDs. Arginine vasopressin is androgen dependent in some brain regions (De Vries & Panzica, 2006), and males are more sensitive to AVP, especially during development. We hypothesize here that AVP, which has a unique role in males, must be present in optimal levels to be protective against ASDs. Either excess AVP or disruptions in the AVP system could play a role in development of the traits found in ASDs. In contrast, OT may possibly be secreted in response to adversity, especially in females, serving as a protective factor.


2008 ◽  
Vol 28 ◽  
pp. 128-149 ◽  
Author(s):  
Inge-Marie Eigsti ◽  
Jillian M. Schuh

As a neurodevelopmental disorder, autism is characterized by impairments and differences at the levels of both brain and behavior. Communicative impairments in autism are a core feature of the disorder, and a rapidly expanding literature is exploring language in autism using the tools of cognitive neuroscience, particularly electroencephalography and brain imaging. Recent research indicates consistent differences in the degree to which language-specific processes are lateralized in the brain, and it also suggests that language impairments are linked to differences in brain structure that may lead to inefficient coordination of activity between different neural assemblies to achieve a complex cognitive task, defined as functional connectivity. We review findings from current work and suggest that neurobiological data are critical in our ability to understand the mechanisms underlying behavioral differences in communicative skills. Going beyond simple dichotomies between delayed versus deviant development, we can use such data to ask whether behavior reflects processes that are merely inefficient or, instead, whether impairments at the behavioral level reflect fundamental differences in brain organization and the networks involved in various tasks.


2021 ◽  
Vol 12 ◽  
Author(s):  
Annefleur M. Koopen ◽  
Eduardo L. Almeida ◽  
Ilias Attaye ◽  
Julia J. Witjes ◽  
Elena Rampanelli ◽  
...  

BackgroundRecent studies demonstrate that a Mediterranean diet has beneficial metabolic effects in metabolic syndrome subjects. Since we have shown that fecal microbiota transplantation (FMT) from lean donors exerts beneficial effects on insulin sensitivity, in the present trial, we investigated the potential synergistic effects on insulin sensitivity of combining a Mediterranean diet with donor FMT in subjects with metabolic syndrome.DesignTwenty-four male subjects with metabolic syndrome were put on a Mediterranean diet and after a 2-week run-in phase, the subjects were randomized to either lean donor (n = 12) or autologous (n = 12) FMT. Changes in the gut microbiota composition and bacterial strain engraftment after the 2-week dietary regimens and 6 weeks post-FMT were the primary endpoints. The secondary objectives were changes in glucose fluxes (both hepatic and peripheral insulin sensitivity), postprandial plasma incretin (GLP-1) levels, subcutaneous adipose tissue inflammation, and plasma metabolites.ResultsConsumption of the Mediterranean diet resulted in a reduction in body weight, HOMA-IR, and lipid levels. However, no large synergistic effects of combining the diet with lean donor FMT were seen on the gut microbiota diversity after 6 weeks. Although we did observe changes in specific bacterial species and plasma metabolites, no significant beneficial effects on glucose fluxes, postprandial incretins, or subcutaneous adipose tissue inflammation were detected.ConclusionsIn this small pilot randomized controlled trial, no synergistic beneficial metabolic effects of combining a Mediterranean diet with lean donor FMT on glucose metabolism were achieved. However, we observed engraftment of specific bacterial species. Future trials are warranted to test the combination of other microbial interventions and diets in metabolic syndrome.


2020 ◽  
Vol 14 (10) ◽  
pp. 1074-1080
Author(s):  
Muluneh Ademe

A growing body of literatures showed the interaction of dysbiotic gut with a wide range of disorders, and the clinical use of fecal microbiota transplantation (FMT) shifted from infectious disease to non-communicable disorders. Despite the promising therapeutic benefits of FMT, the exact mechanisms through which fecal recipients benefit from the fecal intervention are not well understood. However, owing to the advantages of having a healthy gut microbiome, possible mechanisms of actions of FMT has been described. On the one hand, through direct ecological competition, FMT may potentially stimulate decolonization of pathogenic microorganisms and increase host resistance to pathogens. Moreover, following dysbiosis, abnormal microbial colonization of the gastrointestinal tract may also cause excessive or dysregulated immune response, resulting in chronic inflam­mation and the development of mucosal lesions. In this regard, repopulating gut microbiome through FMT helps to restore immune function and reduce host damage. On the other hand, FMT helps to restore essential metabolites used for host metabolism, including short-chain fatty acids (SCFA), antimicrobial peptides (AMP), bacteriocins and bile acids. Therefore, in this review, the existing evidences regarding the mechanisms of action, current opportunities and challenges of FMT will be described.


2020 ◽  
Vol 46 (Supplement_1) ◽  
pp. S204-S204
Author(s):  
Melissa Bauman ◽  
Amy Ryan ◽  
Ana-Maria Iosif ◽  
Takeshi Murai ◽  
Tyler Lesh ◽  
...  

Abstract Background Children born to women who experience infection during pregnancy have an increased risk of brain disorders with neurodevelopmental origins, including both schizophrenia (SZ) and autism spectrum disorder (ASD). Rodent models of maternal immune activation (MIA) have identified the maternal immune response as the critical link between maternal infection and aberrant brain and behavior development in offspring. The nonhuman primate MIA model provides an opportunity to maximize the translational utility of this model in a species more closely related to humans. Our previous pilot study found that rhesus monkeys (Macaca mulatta) born to MIA-treated dams developed behavioral abnormalities and increased striatal dopamine during adolescence. Here we present emerging behavioral outcomes from a larger cohort of MIA-treated nonhuman primates. Methods A modified form of the viral mimic, Polyinosinic-polycytidylic acid (PolyIC), was delivered to a new cohort of pregnant rhesus monkeys (N=14) in the late first trimester (gestational days 43, 44, 46) to stimulate a maternal immune response. Control dams received saline injections at the same gestational time points (N=10) or were untreated (N=4). The offspring are undergoing ongoing comprehensive behavioral evaluations paired with longitudinal neuroimaging to quantify the emergence of brain and behavior pathology associated with prenatal maternal immune challenge. Results MIA-treated dams exhibited a strong immune response as indexed by transient increases in sickness behavior, temperature and inflammatory cytokines. Although MIA offspring developed species-typical milestones and showed no overt signs of atypical interactions with mothers or peers early in development, they had significantly smaller gray matter volume in the prefrontal and frontal cortices than control offspring at 6, 12 and 24 months of age (p < 0.05). At 24 months of age, the animals were tested in a reversal learning paradigm that requires a subject to flexibly adjust its behavior when the reward-related contingencies that it has previously learned are reversed. All animals advanced and performed similarly on the training and initial discrimination phases of the test. However, on the first day of the initial reward reversal, the MIA-treated animals more frequently failed to make a choice as compared to controls (Wilcoxon two-sample test p-value = .005). These emerging data suggest that MIA-treated animals exhibit subtle impairments in cognitive processing. Additional assessments social and cognitive development, including non-invasive eye tracking data, will be presented to further explore the impact of MIA on primate behavioral development. Discussion These findings provide new insights into the emergence of brain pathology in MIA-exposed primates and have implications for the developmental pathophysiology of human psychiatric disorders associated with maternal gestational infection.


2020 ◽  
Vol 10 (1) ◽  
pp. 1-13
Author(s):  
Ananda Deepak V ◽  
Dwarakesh B ◽  
Asifa Nikhat M ◽  
Sridhar SK ◽  
Brito Raj S

In this developing scientific and medical world there is a keen curiosity in studying the activity of human gut micro flora in the emerging revolutionary medical process, fecal microbiota transplantation. This process is about the incorporation of prepared fecal matter solution from the donor into the recipient’s gut which is mainly done to restore and alter the microbial content in the gut of the recipient and to attain a therapeutic impact with greater health benefit. FMT, a bacteriotherapy has proved itself by successfully treating the recurrent clostridium difficile infection in the patient whose disease condition can’t be improved by antibiotic regimen. It also exhibits its therapeutic impact in disease conditions like obesity, irritable bowel syndrome (IBS), metabolic syndrome, inflammatory bowel disease (IBD). In recent studies, research scholar implemented this Microbiota transfer therapy (MTT) in patient with Autism spectrum disorders (ASD) and attained the greater health benefits.


2020 ◽  
Vol 10 (11) ◽  
pp. 814
Author(s):  
Emily M. Borsom ◽  
Keehoon Lee ◽  
Emily K. Cope

The human microbiota is composed of trillions of microbial cells inhabiting the oral cavity, skin, gastrointestinal (GI) tract, airways, and reproductive organs. The gut microbiota is composed of dynamic communities of microorganisms that communicate bidirectionally with the brain via cytokines, neurotransmitters, hormones, and secondary metabolites, known as the gut microbiota–brain axis. The gut microbiota–brain axis is suspected to be involved in the development of neurological diseases, including Alzheimer’s disease (AD), Parkinson’s disease, and Autism Spectrum Disorder. AD is an irreversible, neurodegenerative disease of the central nervous system (CNS), characterized by amyloid-β plaques, neurofibrillary tangles, and neuroinflammation. Microglia and astrocytes, the resident immune cells of the CNS, play an integral role in AD development, as neuroinflammation is a driving factor of disease severity. The gut microbiota–brain axis is a novel target for Alzheimer’s disease therapeutics to modulate critical neuroimmune and metabolic pathways. Potential therapeutics include probiotics, prebiotics, fecal microbiota transplantation, and dietary intervention. This review summarizes our current understanding of the role of the gut microbiota–brain axis and neuroinflammation in the onset and development of Alzheimer’s disease, limitations of current research, and potential for gut microbiota–brain axis targeted therapies.


Sign in / Sign up

Export Citation Format

Share Document