Skip to main content
Gut Pathogens logoLink to Gut Pathogens
. 2025 Jun 1;17:38. doi: 10.1186/s13099-025-00696-2

The functional landscape of the appendix microbiome under conditions of health and disease

Md Shahjalal Sagor 1,#, Tarequl Islam 2,#, Noshin Tabassum Tamanna 3, Md Kamrul Islam Bappy 2, Danishuddin 4,, Md Azizul Haque 4,, Maximilian Lackner 5,
PMCID: PMC12128343  PMID: 40452061

Abstract

Traditionally regarded as a vestigial organ, the appendix is now being reevaluated for its significant function in health and nutrition of humans. Serving as a “safe house” for beneficial, desired gut bacteria, the appendix is protected by resilient biofilms that create a secure environment. This makes the appendix a”basin” for gut microbiota (GM), replenishing the microbial population following disruptions from infections, antibiotic use, or inflammatory bowel disease (IBD). Beyond simply hosting bacteria, the appendix has an active role in functions of the immune system. Disruption of the Appendix Microbiome (AM), such as through appendectomy, was found to result in lowered diversity of gut microorganisms and an increased risk of various diseases. The potential therapeutic applications of the AM are a particularly promising area of research. The appendix’s unique microbial environment and its impact on immunity open new avenues for treatments. These include modulating GM to improve cancer treatment outcomes, mitigating IBD, regulating metabolic pathways in obesity and diabetes, influencing neurotransmitter production in neurological disorders, and addressing cardiovascular and autoimmune diseases. This review highlights the appendix’s transformation from a misunderstood organ to a critical component of gut health and immunity. It explores the function of the human appendix as a resilient reservoir for desired microorganisms, and its role in disease progression. Furthermore, it examines the potential therapeutic applications of AM, presenting exciting opportunities for future research and treatment innovations.

Keywords: Appendix microbiome, Gut health, Appendectomy, Neurological disorders, Immune system, Therapeutic potential

Statement of significance

The Appendix and its Microbiome (AM) that were previously thought of as a useless part of the human body have a deep relation with health and diseases. In this review articles, we broadly discuss factors affecting the function and composition of AM, diseases associated with AM dysbiosis, and its therapeutic potential. Since few studies have addressed the health impacts of the AM, this review offers a comprehensive overview, covering nearly all aspects of the relationship between the GM and AM, and provides noteworthy insights into their roles in health and diseases.

Introduction

The human appendix, once thought to be a vestigial organ, has newly attracted interest for its possible function in maintaining healthy gut microbiota [1, 2]. The term "appendix" comes from the Latin word "appendere," which means "to hang upon" [3]. Historically, the appendix was named because it hangs on to the cecum, which resembles a little appendage suspended from the large structure. This etymology reflects the appendix's presumed function as a secondary addition to the gastrointestinal tract [4, 5]. Research suggests that the appendix functions as “sanctuary” for beneficial gut bacteria, protecting them and aiding in their replenishment after disruptions, such as diarrheal illness [6]. This bacterial reservoir may be crucial for restoring gut flora, which is essential for various bodily functions, including immune response and digestion [6, 7].

The appendix has evolved in an independent way across different species, including rodents, primates,, and marsupials [811]. The recurrence of evolution suggests that the appendix has important functions, rather than being a vestigial organ [9]. Research recommends that the appendix has followed an independent evolution in various mammals, which is a clue for its functional role [9, 12]. It appears that the appendix has been present as an element of mammals’ digestive system for a very long time of at least 80 million years [13, 14]. This is a clue for a selective advantage by this organ [12].

In a healthy state, the AM helps keeping a balanced gut flora, which is essential for the overall body function [15]. In the case of a disease, alterations in AM composition can contribute to or exacerbate conditions like acute appendicitis, which is linked to lowered microbial diversity and a surge in pathogenic bacteria [6, 16]. Furthermore, appendix removal (appendectomy) has been linked to changes in the GM and is believed to cause long-term impacts on human health, including an elevated risk for cancer and certain neurological disorders [7, 1720].

The gut-brain axis (GBA) connects the enteric and central nervous systems, involving hormonal, immunological, and neuronal pathways that together regulate gastrointestinal (GI) function, mood, and cognitive processes [21, 22]. It can be considered a bidirectional network of communication. A crucial component of this axis is the microbiome, consisting of trillions of microorganisms in the gastrointestinal (GI) tract that have a profound impact on the host's health and disease states [23]. Emerging studies suggest that the AM may influence the GBA. The GM can directly impact brain function and behavior, potentially affecting neurodevelopmental and psychiatric disorders [24, 25]. Specific microbial species in the AM are involved in neurotransmitter production. For instance, Lactobacillus and Bifidobacterium species produce GABA (gamma-aminobutyric acid), which helps regulate anxiety and stress responses [26]. E. coli produces serotonin, an important molecule [27]. Enterococcus faecalis produces dopamine, essential for reward and motivation pathways in the brain [27]. Clostridium species set free SCFA (short-chain fatty acids) like butyrate. These exhibitanti-inflammatory effects and can influence brain health [28]. As a microbial reservoir, the appendix may serve a vital function in regulating the GBA, particularly during periods of gut microbial imbalance [29]. The production of neurotransmitters by specific microbial species highlights the complex relationship between the gut and brain, with imbalances potentially resulting in neurological disorders. For instance, reduced GABA and serotonin levels are associated with depression and anxiety disorders [26], while alterations in dopamine production are linked to Parkinson's disease [27]. Furthermore, changes in SCFA production due to AM dysbiosis can impact cognitive function and contribute to Alzheimer's disease [28]. Santos et al. have found that stress is a potentiator of symptom severity in these disorders via the brain-gut-microbiota axis. This highlights the significant clinical implications of understanding the interaction between chronic psychological stress, gut microbiota, and the GBA in mental health and digestive diseases [30]. These insights underscore the significance of the AM in maintaining gut-brain communication and highlight its potential therapeutic implications for neurological health.

The precise mechanisms through which the AM interacts with various bodily functions are still being explored. However, it is hypothesized that the appendix may contribute to health by modulating the immune system, as it contains lymphoid tissue that supports the growth of beneficial gut bacteria. It is also likely that the appendix influences health by acting as a reservoir for bacteria that produce neurotransmitters or their precursors, potentially influencing mood and cognitive functions [22, 23, 31].

The AM is gaining recognition to impact health and diseases. Its potential influence on health offers new insights into conditions ranging from GI disorders and cancers to mental health issues. As research continues to resolve the complexities of the microbiome and its relations with the human body, the appendix may no longer be seen as a redundant organ but as a vital contributor to overall well-being.

In this review, the authors discuss factors affecting AM function and composition, diseases associated with AM dysbiosis (a microbial imbalance in the appendix or gut microbiome), and its therapeutic prospects. Throughout the review, the term “GM” was used frequently, as the GM and AM share similarities at the phylum and genus levels, although their composition differs. Since few studies have addressed the health impacts of the AM, this review offers a comprehensive overview, covering nearly all aspects of the relationship between the GM and AM, and it thereby provides valuable insights into their roles in health and diseases.

Methodology

This review thoroughly examines the determinants of AM function and composition, AM dysbiosis disease, and its therapy. An extensive literature search and meta-analysis was conducted with well-established databases and tools such as Web of Science, PubMed, Scopus, and Dimensions AI tools (Fig. 1) [32, 33]. The search was filtered with 2020 to 2024 published studies that examined the appendix, gut microbiome (GM), and AM functions in health. Some specific keywords like “Appendix microbiota”, “Gut microbiota”, “Appendix microbiota and colorectal cancer”, “Appendicitis and inflammatory bowel disease”, and “Appendicitis and neurological disorder” were used to search relevant articles.

Fig. 1.

Fig. 1

Article selection process for review. The flowchart of the approach for selection of research articles was adapted from the PRISMA flow chart by Moher et al. The chart illustrates the systematic identification, screening, and inclusion of articles to be analyzed. It encompasses stages like identification from bibliographic databases, screening based on publication timeline and relevance, eligibility checks through peer reviews, and final inclusion for thorough analysis. The number of articles retrieved, included, and excluded at different stages is stated in parentheses

To ensure the quality and relevance of included studies, specific exclusion and inclusion criteria were also used. The articles were screened based on their research strengths, peer-review status, and relevance to our review focus. Specifically, studies that discussed the function of the appendix in gut immunity and health, explored the impact of the appendix on disease progression, or investigated therapeutic applications of the AM were given importance. Articles that published between the years 2020–2024 received priority to capture the recent trends in the field. Non-peer reviewed studies, non-AM or GM specific articles, published before 2020, and duplicates/preprints were excluded from analysis.

Data extraction involved meticulous review of the abstracts and texts of the included articles to ensure their relevance and quality. Published data were gathered on different themes, including the appendix as a reservoir for defensive bacteria, impact of appendix removal (appendectomy) on diversity of gut microbiota, immunological functions of the appendix, and therapeutic application of the AM in various diseases. The influence of the appendix on disease progression was also a focal point of the analysis.

To judge the effect of AM on public health, a bibliographic analysis of recent research was conducted using VOSviewer (Fig. 2). The analysis was conducted with VOSviewer version 1.6.20 and Dimensions research publication database with 152 published articles in the period from 2020 to 2024 on AM and related diseases [32, 34]. The analysis was done with 100 keywords extracted from the publications and yielded four clusters in key words.. The network shows the geographical and time spread of the research, with teamwork in addressing AM and its impact..The findings of the network analysis and literature review were combined to give an overview of the appendix's role in gut immunity and health. The analysis brought out the transformation of the appendix from a mysterious organ to an essential component of gut health. It addressed the appendix as a healthy reservoir that is strong, the impact of disruption of the appendix on disease occurrence, and the therapeutic value of the AM in optimizing health outcomes.

Fig. 2.

Fig. 2

Bibliometric analysis of AM literature and related diseases research. VOSviewer version 1.6.20 & Dimensions research publication database were employed for bibliographic network analysis, mapping the relationship of key terms in AM research from 2020 to 2024. The analysis includes 152 studies and 100 keywords. The blue-to-red gradient represents the timeline of 2020 to 2024, describing the trend in research interest in recent time. The figure highlights the relationship between key terms in AM research. Central terms include “appendicitis”, “appendectomy”, “IBD”, and their complications

Factors affecting appendix and/or gut microbiome

The composition of AM and its function is impacted by various factors like diet, disease, antibiotic use, and chemotherapy (Fig. 3) [3537]. By knowing these influences, targeted interventions can be developed, such as dietary modifications or probiotic supplementation, to maintain or restore a healthy AM [24, 38]. As research in this field advances, it will provide further insights into the complex interactions between our lifestyle, health, and the microbial communities within us.

Fig. 3.

Fig. 3

Factors influencing am composition and disease consequences caused by appendix dysbiosis. The diagram illustrates the dual aspects of the appendix microbiome in disease and the factors affecting it. The lower side shows the major factors involved in gut dysbiosis, and the upper side shows pathways leading to the physiological outcomes or diseases due to gut dysbiosis

Effect of diet

Diet is known to be one of the decisive inputs that shape the constituents of the AM. A diet rich in fibers, for instance, increases Firmicutes and Bacteroidetes, which are beneficial and ferment dietary fibers and produce SCFAs (short chain fatty acids), crucial for colon health [35, 39]. In the gut, the fermentation of prebiotic carbohydrates like inulin and fructo-oligosaccharides promotes the growth of good bacteria, primarily Bifidobacterium and Lactobacillus species [40]. Conversely, diets high in processed and sugary foods may encourage the growth of harmful bacteria including Clostridium innocuum, Catenibacterium mitsuokai, Enterococcus spp. [41] C. difficile [42] and C. perfringens [43], etc., leading to dysbiosis [44, 45]. A Western-style diet for one month was found to cause a 71% increase in endotoxemia. By contrast, a more health-conscious diet lead to a 31% decrease [40]. Drug taking can significantly influence the diversity of AM. GM of patients with alcoholism was significantly altered vs. sober ones. A study suggests that alcoholism significantly increases the presence of Firmicutes and lowers the abundance of Bacteroidetes [46]. Vitamins B help the proliferation and metabolic reactions of different bacteria. Around 20% gut bacteria produce Vitamin B12 and around 80% of gut bacteria, like Pseudomonas denitrificans, B. megaterium, and Propionibacterium freudenreichi, B. fragilis, P. copri, C. difficile, F. prausnitzii, R. lactaris, B. animalis, B.infantis, B.longum, and F. varium, need vitamin B12 for their metabolic reactions [47, 48]. The decreased proliferation of Prevotella spp., which breaks down plant polysaccharides and produces thiamine, may be caused by a lack of vitamin B1 [47]. A diet devoid of propionic acid, or taking antibiotics which reduce bacteria generating that compound was found to improve the condition of patients with autism [49]. According to recent findings, a ketogenic diet balances out an unbalanced GM. Following a ketogenic diet, there was a rise in Bacteroidetes and a decrease in Proteobacteria at the phylum level. Following the administration of a ketogenic diet for a few days, a drop in Cronobacter was found. Also, an increase in Prevotella, Bifidobacterium, and Bacteroides was observed at the genus level [50].

Effect of diseases

Diseases, particularly those affecting the GI tract, can significantly alter the AM. For example, acute appendicitis is accompanied by a reduction in microbial diversity and an surge in potentially pathogenic bacteria like Parvimonas and Acinetobacter, which may contribute to the inflammation characteristics of the condition [36]. IBDs (inflammatory bowel diseases) have also been linked to changes in the AM, potentially affecting disease progression and outcomes [5153]. Examples are ulcerative colitis (UC) and Crohn’s disease (CD).

Effect of antibiotics

Antibiotic use is known to disrupt the GM, and the AM is no exception. Antibiotics can reduce the diversity of bacterial species, alter their activity and metabolism, and select antibiotic-resistant ones [5457]. For instance, Penicillin can reduce the number of Lactobacilli, Eubacteria, Bifidobacteria, etc., in the gut. Similarly, quinolones (broad-spectrum bacteriocidals) can reduce the number of Enterobacteriaceae, Coprococcus, and Lachnospiraceae (Table 1) [58, 59]. These changes can lead to antibiotic-associated diarrhea and increase the risk of recurrent Clostridioides difficile infections, highlighting the importance of judicious antibiotic use to preserve the health of the AM [60]. Overuse of antibiotics can lead to the overabundance of Desulfovibrio, or common therapy-resistant bacteria, which emit lipopolysaccharides (LPS) [61, 62]. Behaviors resembling ASD (Autism Spectrum Disorder) were developed as a result of fetal exposure to LPS [63].

Table 1.

Effects of various antibiotics on the abundance of AM and/or GM

Antibiotic groups Antibiotics’ name Microbes decreased Microbes increased
Carbapenems

Carbapenem

Meropenem

Ertapenem

Enterobacteria

Streptococci

Clostridia

Bacteroides

Bifidobacteria

Lactobacillus

Eubacteria

Enterococci
Penicillin

Amoxicillin

Clarithromycin

Erythromycin Spiramycin

Pen V

Lactobacilli

Gram + ve aerobic cocci

Bifidobacteria

Lanchnospiraceae

Eubacteria

Enterobacteria

Bacteroidaceae

Lincomycin Clindamycin

Bacteroids

Blautia

Enterobacteriaceae
Cephalosporins

Cefelor

Cefotaxime

Ceftizidine

Cefuroxime

Cefepime

Bacteroides

Bifidobacteria

E. coli

Enterobacteriaceae

Clostridia

Macrolides

Azithromycin

Clarithromycin

Erythromycin

Spiramycin

Actinobacteria

Lanchnospiraceae

Veillonella

Clostrialis

Erysipelotrichaceae

Bacteroides Proteobacteria

Resistant Enterobacteria

Streptococci

Enterococci

Quinolones & Flouroquinolones

Ciprofloxacin

Norfloxacin

Lachnospiraceae

Coprococcus

Enterobacteriaceae

Gram -ve aerobes

Resistant E. coli

Effect of chemotherapy

Chemotherapy, while essential for cancer treatment, can modulate the AM [64]. Chemotherapy reduces both the number and the diversity of bacteria in the gastrointestinal tract and the feces [65]. Previous studies suggest that it can increase Firmicutes and reduce Bacteroidetes [66, 67]. Some studies suggest that chemotherapy may increase the count of different gram-negative pathogenic bacteria, like E. coli and Pseudomonas, and reduce the abundance of different beneficial gram-positive bacteria, like Bifidobacterium and Lactobacillus [65, 6769]. In addition, this modulation is associated with a higherrisk for infections and can impact the effectiveness of the treatment. Youssef et al. reported that chemotherapy and/or radiotherapy significantly increase Lactobacillaceae and Lactobacillus, and lower Bifidobacteriaceae, Ruminiclostridium, Lachnoclostridium, and Oscillibacter [70]. Moreover, Prevotella copri and Bacteroides plebeius were higher in patients under chemotherapy [71]. This dysbiosis consequently impacts the body in different ways [72].

Effect of genetic conditions

Through a complex interplay of host and environmental variables, host genetic variations influence GM [73]. Certain genetic variations are accountable for preserving the gut commensal makeup [74]. Variants at the LCT locus associated with higher levels of Bifidobacterium genus and Negativibacillus genus, UBA3855 sp900316885 and CAG-81 sp000435795. In addition, higher levels of Faecalicatena lactaris are associated with ABO variables. MED13L variants linked to colorectal cancer (CRC) showed higher levels of Enterococcus faecalis [75]. Similarly, PLD1 & LINGO2 variants related to obesity showed a higher number of Akkermansia and Blautia spp. [76], and NOD2 (nucleotide-binding oligomerization domain-2) variants having IBD showed a higher number of Escherichia spp. and a lower number of Faecalibacterium spp. [77]. SLIT3 (slit guidance ligand 3) variants associated with inflammation and obesity showed an increase of Clostridiaceae and Dermococcus spp. [78]. There are some more genes that impact the gut microbial composition. All of these prove a strong association of host genetics with GM.

Appendix and/or gut microbiome in disease development

The AM is highly important both for gut health and immune function. Changes in the AM can lead to diseases such as acute appendicitis, IBD, back pain, obesity, diabetes, cancer, and so on [52, 79, 80]. Figure 4 shows the microbial compositional changes that cause different disease development, whereas Fig. 5 shows the effects of gut dysbiosis in different physiological outcomes.

Fig. 4.

Fig. 4

Involvement of GM in different disease development. The red arrows on the left side indicate the reduction of microbes associated with the disease development and the green arrows on the right side indicate the increase of the microbes associated with the disease development. It is clear from the figure that a reduction in the number of Bifidobacterium, Lactobacillus, Prevotella, Faecalibacterium, and Firmicutes/Bacteroidetes is associated with most of the disease progression. On the other hand, an increase in the number of E. coli, Enterococcus, Akkermansia, Enterobacteriaceae, Ruminococcus, Clostridium, and some other pathogens is associated with most of the disease progression. This could be a piece of important information for microbe-mediated therapeutics

Fig. 5.

Fig. 5

Effects of gut dysbiosis in different physiological outcomes. The left side of the figure shows the effects on different organs resulting from the dysbiosis of GM. The right side of the figure shows the pathways along the body that are involved in different physiological outcomes or diseases due to the dysbiosis of GM. GABA gamma-aminobutyric acid, IL interleukin, TNF tumor necrosis factor, HPA hypothalamic–pituitary–adrenal axis

Acute appendicitis

Globally, acute appendicitis ranks top among surgical emergencies. The conventional theory for its pathogenesis has been luminal obstruction, but emerging evidence suggests a strong involvement by the microbiome in the development of this condition [6]. E. coli, Streptococcus anginosus, P. aeruginso, Bacteroides spp., and Klebsiella spp. are common bacteria that can cause acute appendicitis [81, 82]. Studies have shown that patients with acute appendicitis exhibit a significant decrease in microbial diversity compared to healthy individuals. Parvimonas and Acinetobacter are increased, whileFaecalibacterium, Blautia, and Lachnospiraceae (commensal taxa) [6] are reduced. Fusobacterium spp., Porphyromonas, and Parvimonas can be translocated from the mouth to the appendix, and these pathogeens can increase the severity of complicated appendicitis [83]. Other studies found a huge number of Fusobacterium spp. in the saliva and feces of children suffering from pediatric acute appendicitis [84, 85]. 16S rRNA gene sequencing of the appendiceal microbiome revealed that Campylobacter jejuni may be a significant cause of acute appendicitis [86]. These changes in the microbiome may result in inflammations, resulting in acute appendicitis.

Inflammatory bowel diseases (IBDs)

IBDs are connected with severe inflammation of the GI tract. The appendix has been found to be involved in the pathogenesis and outcomes of IBDs [87, 88]. Alterations in the GM, such as decreasing the number of Actinobacteria, Firmicutes, and Bacteroidetes, and enrichment of Proteobacteria, could start the disease [79, 88]. The study revealed that patients with BD had higher concentrations of Bilophila spp., a sulfate-reducing bacteria (SRB), and several opportunistic pathogens, such as Parabacteroides spp. and Paraprevotella spp. Additionally, there was a lower concentration of methanogens, such as Methanoculleus spp. and Methanomethylophilus spp., and butyrate-producing bacteria (BPB) Clostridium spp. [89]. Recent research has highlighted significant microbiome alterations in Crohn’s patients, with the detection of the Staphylococcus sciuri pathogen and elevated NLRP3 protein expression, suggesting its potential as a diagnostic biomarker [90]. For instance, inflammation around the appendix, known as the “peri-appendicular patch”, has been frequently observed in patients with UC, even preceding the onset of the disease [88]. Moreover, the temperate phage viruses residing in the bacterial population display a shift from lysogenic to lytic replication in IBD patients [91]. Alterations of fungal and protozoa populations were also observed with IBD. Patients with IBD had a higher frequency of fungi, mostly Saccharomyces cerevisiae, and a lower frequency of protozoa, mostly Blastocystis species (subtypes 1, 2, 3, and 4) [92].

Auto-immune disease

Gut dysbiosis has a strong relationship to the development of ADs like RA, type 1 diabetes, systemic lupus erythematosus, multiple sclerosis, spondyloarthritis, and irritable bowel syndrome [80]. The abnormal generation of autoantibodies is a characteristic of autoimmune disorders. The immune system is influenced by genetic and/or environmental variables, which result in aberrant production of proinflammatory cytokines and the decline of anti-inflammatory cytokines, autoreactive T cells, and B cells that produce autoantibodies. The rising prevalence of autoimmune disorders may be caused by significant changes in the GM as a result of dietary modifications, compromised gut integrity, extensive use of antibiotics, and other factors [68, 80, 93]. Molecular mimicry, effects on intestinal mucosa permeability, the host immunological response triggered by the microbiota, antigenic mimicry, and epitope spreading are among the potential pathways leading to AD (Fig. 6).

Fig. 6.

Fig. 6

Role of microorganisms in autoimmune disease. The diagram illustrates the interactions between gut pathogens, commensal antigens, microbial antigens, and immune cells, highlighting the complex pathways involved in gut immunity and potential therapeutic targets in gut-related diseases

An important player in the immune response is the aryl hydrocarbon receptor AhR, a ligand-activated transcription factor that detects substances found in food, secondary metabolites produced by microbes, and environmental contaminants to regulate transcriptional programs in the immune system. Gut barrier development and maintenance depend heavily on AhR [94]. AhR may be involved in autoimmune illnesses through the translation of extrinsic and intrinsic signals into cellular responses through its binding to a variety of ligands coming from diet, cells, and microbes [95]. In AhR-deficient animals, reduced levels of innate IL-22 facilitated the growth of immunological activator commensal segmented filamentous bacteria (SFB), which in turn promoted Th17 cell proliferation. By inhibiting pathogenic Th17 cells, the innate expression of AhR reduced the impact [96]. The AhR ligand 2,3,7,8-tetrachloro dibenzo-p-dioxin (TCDD) caused changes in the abundances of SFB and the immunological suppressor Bacteroides fragilis in mice. Furthermore, the GM's presence of SFB considerably controlled the TCDD-induced host response, indicating the therapeutic potential of AhR ligands and important commensals [97].

Another cause of autoimmune illness is dysbiosis. The GMs molecularly mediate autoimmune through cross-reactivity and posttranslational modification of autoantigens. Live gut bacteria can translocate across a malfunctioning gut barrier, causing direct interactions with tissue and immune cells that in turn trigger systemic autoimmunity at the cellular level [98]. Research conducted both in vitro and in vivo has demonstrated that dysbiosis in patients with systemic lupus erythematosus (SLE) can enhance Th17 differentiation and lymphocyte activation. Conversely, Bifidobacterium bifidum supplementation can balance the Treg/Th17/Th1 ratio and prevent CD4 + lymphocytes from being overactivated [99]. Additionally, serum antibodies against Ruminococcus antigens and decreased bacterial diversity with a five-fold increase in R. gnavus prevalence in SLE patients point to the role of GM dysbiosis in the pathophysiology of lupus nephritis [100]. Similarly, autoantibody levels and the duration of RA were linked to low gut microbial diversity and high Collinsella abundance, which was coupled with the pro-inflammatory cytokine IL-17A. This shows that these variables may be used to predict the status of RA disease [101]. Additionally, compared to ACPA-negative persons, RA patients with positive tests for the anti-citrullinated protein antibody (ACPA) showed a lower level of microbial diversity and enrichment of Blautia, Akkermansia, and Clostridiales [102].

There are a number of potential explanations for the correlation between dysbiosis of the GM and autoimmune disorders, including effects on the functioning of the human immune system. For example, altering the host immune response and stimulating antigen-presenting cells (APCs), such as dendritic cells (DCs), may cause antigen presentation and the release of cytokines, which in turn may impact the development and functionality of T cells. Moreover, this effect throws off the balance between T regulatory cells (Tregs) and T helper 17 (Th17) cells. Antigenic mimicry has been linked to similarities between foreign and self-antigens; as a result, pathogen-derived autoreactive T and B cells are activated, which in turn promotes autoimmunity [93]. Some microbes can change the host antigen using their own enzyme-producing neoepitopes which are detected by the immune system causing autoimmune disease. A greater abundance of Prevotella copri has been linked to higher protein citrullination which subsequently stimulates pro-inflammatory cytokines, leading to damaged tissues, and self-antigens release, which supports to the development of autoimmune responses [103].

Cancer

Appendicitis can be a precursor to appendix cancer. Tumors within the appendix can block the organ, leading to the trapping and overgrowth of bacteria normally present in the intestines. This bacterial overgrowth may contribute to inflammation and other changes that could eventually lead to cancer [104]. While R&D has concentrated on the relationship between bacteria and appendix health, there is currently limited understanding of how intestinal fungi may be involved [7, 105]. Mechanisms by which AM influences cancer risk include immune modulation, production of carcinogenic metabolites, and direct interactions with mucosal cells (Table 2) [106, 107]. Emerging evidence underscores the crucial role of gut microbes in CRC development [17, 108]. Recent studies have proposed the involvement of Fusobacterium nucleaturm, Pseudomonas, Streptococcus bovis, Salmonella, Helicobacter pylori, Akkermansia, Bacteroides fragilis, Peptostreptococcus, and Clostridium septicum, in the CRC progression [80, 109]. Metabolites of these bacteria like bile salts, hydrogen sulfide, and trimethylamine-N-oxide (TMAO) cause inflammation, DNA damage, and subsequently cancer development [109]. Observational studies and clinical trials suggest an association between GM and cancer. Recent studies, including a two-sample Mendelian randomization study, have investigated the causal link between GM and cancer. These studies identified multiple associations between genetic liability in the GM including the genus Bifidobacterium and cancer, emphasizing the microbiota's causal role in cancer development [110, 111]. Pancreatic ductal adenocarcinoma (PDAC) is one of the fatal solid tumors which is resistant to immunotherapy. Metagenome analysis of the AM of PDAC patients showed an abundance of Klebsiella pneumoniae, Bifidobacterium animalis, and Adlercreutzia equolifaciens, while certain commensals are depleted [112]. Additionally, identifying microbial signatures associated with cancer may lead to novel biomarkers for early detection [113, 114].

Table 2.

Gut-microbial involvement in different types of cancer progression

Domain Microorganisms Metabolites Cancer type Mode of action References
Bacteria Fusobacterium nucleatum Fusobacterium adhesion protein (FadA) CRC (colorectal) Promotes adhesion and invasion of cancerous cells [115, 116]
E. coli Colibactin, Cytolethal distending toxin (CDT), Genotoxin, Various cancers (including colorectal, breast, and pancreatic)

Genotoxicity.

Disrupts cell cycle and promotes DNA damage

[117, 118]
Peptostreptococcus spp. Hydrogen sulfide CRC Promotes inflammation and genotoxicity [119]
Helicobacter pylori Cytotoxin-associated gene A (CagA) Gastric Cancer Chronic inflammation induced by the bacteria can damage the stomach lining [116, 120, 121]
Parvimonas micra LPS CRC May induce chronic inflammation and immune suppression [122, 123]
Enterococcus faecalis Unidentified CRC Increase the frequency of aneuploidy and tetraploidy [124]
Sulfidogenic Bacteria (i.e. FusobacteriumDesulfovibrio, Bilophila wadsworthia) Hydrogen sulfide (H2S) CRC DNA damage, Effect DNA repair, Chromosomal instability (CIN), and Interference with mitochondrial function [116]
Bacteroides fragilis B. fragilis toxin (BFT) CRC May contribute to tumor growth and metastasis [125]
Akkermansia muciniphila (Possible protective effect) Short-chain fatty acids (SCFAs) Various cancers May enhance anti-tumor immunity and reduce inflammation [126]
Fungi Candida albicans Various GI cancers May promote inflammation, reduce cell adhesion (facilitating metastasis), and influence tumor gene expression [127]
Malassezia spp. Possibly unidentified metabolites Breast cancer Potential role in promoting inflammation and reducing survival rates. More research is needed [128]
Archaea Methanobrevibacter spp. Methane CRC Disruption of gut barrier function and inflammation [129]
Methanothermobacter spp. CRC (indirect—via IBD) Modulation of gut immune response (linked to IBD) [130]
Thermoplasmatales CRC Unclear, potentially linked to gut inflammation [131]
Candidatus Mancarchaeum acidiphilum CRC (potential biomarker) Correlated with other cancer-associated microbes, function unknown [131]

Obesity

An increasing amount of data is available on modifications to microbiota and obesity, along with the illnesses linked to it. Age-related, exercise-related, and weight-related alterations have all been documented [132]. The intestinal microbiota influences the development of obesity [133, 134].

Conversely, individuals with high blood pressure and resistance to insulin had lower bacterial richness. Additionally, distinct correlations were discovered between elevated pressure and the existence of Clostridium and Clostridiaceae, reduced low-density lipoprotein (LDL) and Lachnospiraceae, Gemellaceae, Turicibacter, and elevated risk of metabolic syndrome [135]. Similarly, intestinal abundance of Akkermansia muciniphila has been observed to be negatively correlated with the probability of greater severity of insulin resistance in both lean and obese Asian patients [136].

The effects of GM on other metabolic processes linked to immune functions may also have an impact on bodyweight control. This is primarily because GM has been linked to elevated levels of bacterial LPS, which have been found in subjects whose obesity was brought on by a high-fat diet [137]. Increased intestinal porosity, an immunological response in the mucosa, and increased intestinal permeability are all caused by dysbiosis. It seems that the microbiota facilitates the transfer of LPS but also releases it, which might result in insulin blockage and metabolic endotoxemia [132, 137]. Moreover, research shows the ratio of Firmicutes/Bacteroides in the obese gut [138]. In another study, infants with normal weight showed a higher richness of Lactobacillus, whereas Rhomboutsia and Clostridium sensu stricto predominated in the obese cohort [135].

Diabetes

An autoimmune response is thought to cause Type 1 diabetes. The beta cells in the pancreas, which produce insulin, are destroyed by this process. Type 1 diabetes patients were found with a reduced abundance of Bifidobacterium, Lactobacillus, Faecalibacterium, Firmicutes/Bacteroidetes ratio, and a greater abundance of Bacteroides and Bacteroidetes [80]. Another study on gnotobiotic mice (born in germ-free conditions and inoculated with known bacteria) discovered that the intestinal bacteria impacted glucose metabolism raised the accumulation of macrophages toward the pro-inflammatory M1 phenotype in white adipose tissue, and ultimately led to the formation of insulin resistance. This finding established a link between bacterial LPS and insulin resistance [139]. The existence of Coriobacteriaceae (Faecalibacterium prausnitzii), Parabacterodes, Bacteroides caccae, Oscillospira, Parabacteroides distasonis, Coprococcus, and Haemophilus parainfluenzae was reported to be linked to a reduced metabolic syndrome score, fasting glucose, and HOMA_IR (Homeostatic Model Assessment for Insulin Resistance) [135].

Rheumatoid arthritis

It has been shown that the bacterial makeup of the intestinal tract differs in patients with rheumatoid arthritis (RA) compared to healthy individuals. Reduced numbers of Firmicutes, Bacteroides, Bifidobacterium, F. prausnitzii, Haemophilus spp., and Roseburia were observed in RA patients, while increased numbers of Prevotella copri, P. gingivalis, Collinsella, Lactobacillus, Bacteroidaceae, Lachnospiraceae, Bacteroidetes, Clostridiaceae, and L. salivarius were observed [80, 140142]. RA caused by GM is mainly a result of an autoimmune response [140, 142]. Dysbiosis triggers autoreactive T cells in the colon, which enhances susceptibility to arthritis. Dysbiotic microbiota in the colon trigger autoreactive SKG mice T cells, which results in inflammation of the joints [142]. P. copri dominated the GM of patients with early-stage RA. When given zymosan, SKG mice with microbiota from RA patients experienced severe arthritis and an increase in intestinal Th17 cells [140, 142]. In individuals with new-onset untreated rheumatoid arthritis (NORA), increases in Prevotella abundance were associated with decreases in Bacteroides and a loss of microorganisms that were thought to be helpful. An elevated susceptibility to chemically induced colitis was another effect of P. copri [103].

Role of appendix and/or gut microbiome in neurological disorders

The GBA is an intricate bidirectional communication network that links the GI tract with the central nervous system (CNS). This dynamic interplay involves complex interactions through neural, immune, and endocrine pathways [143, 144].

The GM contributes to CNS homeostasis by modulating the immune system and regulating the production of various molecules and metabolites [38, 145]. These metabolites, such as SCFAs produced by Bacteroidetes and Firmicutes, neurotransmitters, and other signaling molecules, can cross the blood–brain barrier and impact brain function directly [146, 147]. Moreover, the immune system's interaction with the GM helps maintain a balanced inflammatory response, which is crucial for preventing neuroinflammation [148, 149].

Neurological disorders (NDs) such as depression, schizophrenia, Parkinson disease (PD), Amyotrophic lateral sclerosis (ALS), autism spectrum disorder (ASD), epilepsy, and migraine have been linked to dysbiosis in GM [150]. While working with the C57BL/6 J mouse model, depletion of Lactobacillus and enrichment of Akkermansia were associated with neuroinflammation [151]. According to a recent study, stress may have caused gut dysbiosis, which in turn may have increased the abundance of conditional pathogens like Enterococcus, Vagococcus, and Aerococcus, as well as pathogens like E. coli and Shigella [152]. Alcoholism patients' GM differed significantly from healthy adult patients' GM. Firmicutes greatly increased in number whereas Bacteroidetes significantly declined. Mice receiving fecal microbiota from alcoholism patients displayed behaviors resembling anxiety and depression, a reduction in socialization, a spontaneous favor for alcohol, and a reduction in brain-derived neurotrophic factor (BDNF) when compared to mice transplanted with fecal microbiota from healthy male adults [46]. It's interesting to note that individuals with schizophrenia typically experience GI complications, have a greater frequency of intestinal barrier failure, and exhibit higher bacterial translocation [153]. The genera like Succinivibrio, Megasphaera, Collinsella, Clostridium, Klebsiella, and Methanobrevibacter were significantly higher in schizophrenia, whereas Blautia, Coprococcus, and Roseburia were abundance in healthy controls. Patients with first experience of schizophrenia significantly lowered numbers of Bifidobacterium, E. coli, and Lactobacillus [154]. A similar result was reported by other studies that showed a correlation between Succinivibrio and Corynebacterium and the degree of schizophrenia symptoms [155]. Another study reported an increase in Anaerococcus and a decrease in Haemophilus, Sutterella, and Clostridium in schizophrenia [156].

PD showsdegeneration of dopaminergic neurons. GM alterations may precede the onset of motor symptoms in PD. Interventions aimed at restoring microbial balance could potentially slow down disease progression [157, 158]. Increased genus Akkermansia, which promotes intestinal permeability, and decreased SCFA-producing bacteria of the genera Roseburia and Faecalibacterium are frequently linked to PD [159]. The most consistent change in the GM composition in PD was found in a more recent meta-analysis of eleven case–control studies, which revealed decreased genus Faecalibacterium and Lachnospiraceae family and increased genera Lactobacillus, Akkermansia, and Bifidobacterium [160]. Shen et al. reported similar changes in a meta-analysis involving fourteen studies. Reductions in Prevotellaceae, Faecalibacterium, and Lachnospiraceae were observed in the analysis, whereas increases in Bifidobacteriaceae, Ruminococcaceae, Verrucomicrobiaceae, and Christensenellaceae were noted [161]. Rodent models of PD have shown a similar pattern of gut dysbiosis [162]. Conflicting findings regarding the role of the appendix and PD been reported by recent studies. Park et al. found no link between appendectomy and risk change of PD in the Korean population, indicating that appendectomy did not determine the risk of PD [163]. In contrast, Wang et al. found out that appendectomy is associated with a lowered risk of PD, highlighting a potential protective effect of the surgical removal of the appendix [164]. Similarly, Jain et al. found no increased risk of PD following appendectomy, suggesting that the appendix is not a critical site for the development of PD pathology [165].Nakahara et al. described a significant phylogenetic difference in the gut microbiota between PD patients and healthy controls who had undergone appendectomy [166]. Their findings suggested a potential role of the gut microbiota, in particular the family Enterobacteriaceae, in the development of PD. The variable findings of these studies point to a variety of limitations and areas for future research. The studies were conducted in different populations, and this may be the reason behind the variable results. The studies were conducted through different methods, such as Cox regression analyses and Mendelian randomization [163, 164, 166], which may influence the results. Genetic susceptibility to PD may vary between different populations, and this might influence the relationship between appendectomy and risk for PD. Potential confounders such as age, sex, and underlying illness may not have been accounted for in all of the studies. Larger populations and more representative groups are needed to validate the results and a clearer causal link. Some studies, like Nakahara et al. are preliminary and must be validated by larger, more precise studies [166]. To bridge these gaps, subsequent studies should involve large-scale, multi-center studies with heterogeneous populations to account for both genetic as well as environmental heterogeneity. Standardized methods between studies would allow better comparison of outcomes. In addition, control of potential confounders is important to differentiate the true effect of appendectomy on the risk of PD. Long-term follow-up studies are required to ascertain the timing of the relationship between appendectomy and pathogenesis of PD. Mechanistic investigations should also study the precise mechanisms by which appendix and gut microbiota affect PD pathology.

ALS involves the degeneration of motor neurons. While less is known about the GM's role in ALS compared to AD and PD [167]. A study with ALS SOD1G93A mice yielded a reduction in Butyrivibrio fibrisolvens [162], which liberate butyrate. However, while working with humans no substantial changes were found in the ratio of Firmicutes:Bacteroidetes [168].

ASD is a collection of neurodevelopmental disorders. Concomitant pathologies in patients with ASD typically include gut dysbiosis, anxiety, depression, seizures, and other GI issues, like diarrhea, abdominal discomfort, and vomiting [169, 170]. It has been observed that people with ASD have higher biomass and less variation in bacteria [171, 172]. The most significant might be lower concentrations of Bacteroidetes, Actinobacteria, Proteobacteria, Prevotella, Corprococcus, and Veilonellaceae [171], combined with higher concentrations of Desulfovibrio spp., Sutterella spp., and Ruminococcus torques, or Clostridium spp. [173, 174]. Additionally, other authors reported an excess of Megamonas, and Candida spp., which were found to set freeammonia and other toxins that contribute to autistic behavior. They also saw a decline in the ratio Bacteroidetes:Firmicutes [174176]. Additionally, the onset of Asperger's syndrome was connected with a decrease of pro-inflammatory in Bifidobacterium spp. [177]. According to earlier research, patients with autism may benefit from butyrate-producing bacteria like Eubacterium, Ruminococcaceae, Erysipelotrichaceae, and Lachnospiraceae [178].

Epilepsy is a seizure disorder and aneurological disease. GM composition differs between drug-resistance and drug-responsive epilepsy patients. Compared to patients who experienced more than four seizures annually, those who experienced four or fewer per year displayed higher levels of Bifidobacteria and Lactobacillus [179]. Recent studies suggest a ketogenic diet to restore GM and protest epilepsy. The level of Parabacteroides and Akkermansia muciniphila was elevated by the ketogenic diet. Seizures were prevented when gnotobiotic cocolonization was enhanced with Akkermansia and Parabacteroides [180]. Moreover, probiotic supplementation of Lectobacillus and Befidobacterium reduced 50% of drug-resistance epilepsy [181].

A “leaky gut”-related increase in proinflammatory cytokines may contribute to the onset of migraine discomfort [182]. A significant correlation between GM and migraine was established. There was a notable enrichment of Firmicutes, particularly Clostridium spp., in the migraine group, whereas, the healthy controls had higher concentrations of advantageous bacteria, including Methanobrevibacter smithii, Bifidobacterium adolescentis, and Faecalibacterium prausnitzii [183].

Role of appendix and/ or gut microbiome in parasitological disorders

The microbiome in the gut plays a very crucial role in protecting the host organism against numerous infections, including those caused by parasitic protozoa and fungi. The intestinal microbes regulate fat storage, stimulate epithelial cells, and influence brain and immune system development. All these interactions are significant in maintaining homeostasis and preventing infections [184, 185]. Intestinal protozoa-microbiome interactions emphasize the immunomodulatory effect of the intestinal microbiota on protozoan infection like Entamoeba histolytica, Blastocystis spp., Giardia duodenalis, Toxoplasma gondii, and Cryptosporidium parvum [184, 185]. The organization of intestinal bacterial communities can modulate the path of protozoan infection and the destiny of parasitic disease [186]. In parasitic illnesses associated with intestinal parasites, e.g. amoebic colitis or amoebic liver abscess due to E. histolytica or intestinal giardiasis due to G. duodenalis, interactions between intestinal bacteria and such parasites could affect parasitic or bacterial virulence. For instance, reduced abundance of Bacteroides, Clostridium, and Lactobacillus and increased abundance of Bifidobacterium is associated with amoebic colitis, while increased abundance of Prevotella copri is associated with amoebic diarrhea [187, 188]. Non-pathogenic parasitic protozoa also have the potential to alter intestinal microbiota diversity and provoke host immune responses. For example, asymptomatic colonization with Blastocystis spp. is associated with raised total diversity of intestinal bacteria and widespread changes of the dominant species within the gut microbiota. But colonization by Blastocystis hominis has been associated with reduced numbers of some markers of intestinal immunity, suggesting an anti-inflammatory environment within the intestine [184, 189, 190].

Current investigations explore the potential of fecal microbiota transplantation (FMT) to control immune response. There is evidence that FMT from protozoa-exposed donors can inhibit the immune response in a germ-free mouse model, demonstrating its potential to modulate immune responses and influence intestinal physiology [184, 188]. Specifically, gut protozoa such as Blastocystis hominis and Entamoeba histolytica can modulate the immunoinflammatory status and induce functional changes in the intestine by interacting with the gut microbiota [188, 191]. This downregulation of intestinal immune response includes decreased production of such proinflammatory cytokines as IL-6, TNF, IFN-γ, MCP-1, IL-10, and IL-12 in FMT-receiving mice from protozoa-exposed donor mice [188, 192]. Investigation into intelligent biological networks indicates a potential role through increasing anti-microbial resistance resilience by nutritional interventions. Enteric protozoan pathogenic infection effect on global health and the gut microbiome contribution towards infection is high-profile. Nutritional supplementation, including proteins, prebiotics, probiotics, synbiotics, and food formulations, is discussed as being a key part of recovery from dysbiosis and resistance to opportunistic pathogens [193]. Systems biology strategies are valuable tools to define the biochemical pathways of infection and recovery, allowing for better understanding of the impact of nutritional treatments on the gut microbiome and its role in protozoal infections [193, 194].

The imbalance of the gut microbiota, known as dysbiosis, can significantly impact the health status of the host. For example, Cryptosporidium spp. is an epithelial-lining-bound parasite that interacts with bacterial biofilms in the lining of the gut to cause behavioral adaptation in itself and neighboring bacterial populations [193, 195]. Studies have shown that Cryptosporidium growth is directly proportional to Pseudomonas aeruginosa biofilm maturation, indicating the complex interaction between parasites and bacterial communities [193, 195]. Similarly, Giardia infection may provoke alterations in gut microbiota, converting commensal bacteria into pathogenic forms and leading to increased virulence and inflammation [196]. In addition, protozoan infections can produce cross-organ effects. For instance, Cryptosporidium and Entamoeba have been known to cause opportunistic bronchial and pulmonary invasion, leading to respiratory conditions such as pneumonia, cough, and chest pain [193, 197]. Infections of the enteric type can lead to the host immune system releasing cytokines, which in turn cause inflammation and asthma. Exaggerated production of short-chain fatty acids (SCFAs) during infection potentially controls the immune system and dampens allergic responses, permitting regulatory T-cell differentiation [198]. These experiments highlight the interplay between the appendix, gut microbiota, and parasitological diseases. Microbiota manipulation has the potential to offer novel therapeutic approaches to the management of intestinal protozoan infections and related immune responses.

Appendix microbiome as a possible therapeutic approach

The AM may help to prevent and treat various diseases and could be leveraged for therapeutic approaches. They could be used to treat cancer, GI diseases, metabolic disorders, neurological disorders, cardiovascular diseases (CVDs), and so on [60, 115, 199201]. Methods that were applied to treat these diseases using appendix/GM are the use of prebiotics, probiotics, FMT (fecal microbial transplantation), engineered bacteria, phage therapy, RNA-guided gene therapy, and diet and metabolic modification (Fig. 7) [60, 199, 202205]. The main purpose of these methods is to reform the GM which ultimately impacts the overall health outcome.

Fig. 7.

Fig. 7

Appendix microbiome as possible therapeutic. The figure shows different possible methods for reforming gut dysbiosis, treating diseases, controlling pathogens, and inhibiting resistance or virulence genes

Prebiotics, probiotics, and postbiotics

Prebiotics are food ingredients that help the growth of helpful gut bacteria and thereby benefit the host. There are different types of prebiotics like fructans, galacto-oligosaccharides (promotes the growth of Bifidobacteria, Lactobacilli, Enterobacteria, Bacteroidetes, and Firmicutes), starch and glucose-derived oligosaccharides (promotes the growth of Bifidobacteria, and Firmicutes) [206]. Probiotics are beneficial bacteria that keep the gut healthy. Recent studies suggest that probiotics could serve as an adjunctive therapy in acute appendicitis, hinting at the broader therapeutic potential of targeting the AM. By influencing the immune response and possibly restoring a healthy microbial balance, probiotics might offer a non-surgical alternative to appendectomy in uncomplicated cases of appendicitis [2, 204].

Probiotics influence the efficacy and toxicity of cancer treatments [6, 115]. Probiotics, such as Lactobacillus casei supplements, have shown promise in reducing chemotherapy-induced diarrhea, a common side effect [207]. Over the past ten years, Akkermansia muciniphila, a representative commensal bacterium, has drawn a lot of interest as an effective Next-generation probiotic (NGP) that can influence tumorigenesis either directly or indirectly. It does this, in part, through its effects on antitumor immunosurveillance, which includes stimulating pattern recognition receptors (PRRs) and improving outcomes of GI cancer [208].

Probiotics also serve as a promising solution in reducing the severity of neurological disorders. Previous results suggest that treating CD with Faecalibacterium prausnitzii as a probiotic may be a promising approach for preventing dysbiosis [209]. In a clinical trial, patients with drug-resistant epilepsy benefited from a 4-month probiotic regimen that included L. acidophilus, L. plantarum, L. casei, L. helveticus, L. brevis, B. lactis, and Streptococcus salivarius subsp. Thermophilus. This regimen resulted in a ≥ 50% drop in the number of seizures in 28.9% of the patients [181]. A mother’s GM needs to be healthy and balanced during pregnancy to maintain a healthy and balanced milk microbiota composition [210]. Furthermore, oral probiotic administration may enhance the quantity of Bifidobacterium and Lactobacilli in breast milk in moms who give birth vaginally [210].

Postbiotics are bioactive compounds produced by probiotics upon digestion of prebiotics. Using postbiotics is a unique strategy for treating disorders like CVD and gut dysbiosis at the same time. They are described as the creation of inactive microbes and/or their components that benefit the host's health [211]. The connections between gut microbes and brain cells, such as microglia, astrocytes, and neuronal cells, are impacted differently by diet-derived gut microbial metabolites. Methods to alter microbial metabolite levels have been proposed as a target for neuropsychiatric disorders [212, 213]. Colonocytes and a number of other cell types respond positively to butyrate in vitro. Butyrate reduced the expression of vascular cell adhesion molecule-1 (VCAM-1) and intracellular cell adhesion molecule-1 (ICAM-1) caused by TNFα. IL-1-stimulated VCAM-1 and ICAM-1 expression was suppressed by butyrate at 10 mmol/L [214]. When sodium acetate (1 g/kg) is given intraperitoneally into radiotelemetry-implanted mice, acetate reduces both the heart rate (HR) and the mean arterial pressure [215]. Propionic acid, a molecule generated from microbiota, has shown remarkable efficacy in the metabolism of intestinal cholesterol. After taking 500 mg twice a day for eight weeks, hypercholesterolemic patients showed reductions in total cholesterol, very-low-density lipoprotein (VLDL), and low-density lipoprotein (LDL) [216].

Diet and metabolites

Eating a balanced and healthy diet and its microbe-derived metabolites is an excellent solution for GM-mediated therapeutics for different diseases. In vivo, study shows that exogenous Lactobacillus reuteri inhibits the growth of colon tumors, increases tumor reactive oxygen species, and reduces translation of proteins. As shown by microbial profiling, L. reuteri and its metabolite, reuterin are downregulated in human and mouse CRC [217]. In female mice, estrogens can slow the evolution of CRC by increasing the colonic expression of SLC3A2, which binds to the D-alanyl-D-alanine carboxypeptidase (DD-CPase) of Carnobacterium maltaromaticum [218].

The unique bacterial populations in the appendix may influence metabolic pathways and gut hormone production. AM could regulate the production of SCFAs, which are important for insulin sensitivity and energy balance [219221]. Certain appendix bacteria (i.e. Proteobacteria, Bacteroidetes) could produce or influence the production of neurotransmitters such as serotonin and gamma-aminobutyric acid (GABA) [222, 223]. These compounds are known to control the mood.

The GM’s impact on CVD is multifaceted, affecting systemic inflammation, cholesterol metabolism, and blood pressure all vital in CVD development [224226]. Dysbiosis can lead to inflammation and compromised intestinal barriers, increasing CVD risk. Microbial metabolites like trimethylamine N-oxide (TMAO), linked to atherosclerosis, are produced by gut bacteria (i.e. Acinetobacter, Pelobacter) from dietary nutrients [227]. Additionally, GM (i.e. Bacteroides sp, Clostridium, Eubacterium) influences bile acid metabolism, affecting cholesterol levels and CVD risk [201, 228, 229].

Fecal microbial transplantation (FMT)

FMT has shown promise in restoring GM balance after certain cancer treatments [202]. Additionally, the emerging field of pharmacomicrobiomics explores how GM influences an individual's response to medications. By analyzing GM, doctors may give a prediction of patient response to cancer treatments, paving the way for personalized medicine [230]. FMT has shown promise in treating IBD [231]. Therapeutically, there is potential for using appendix-derived microbiota in FMT for patients with metabolic disorders [199]. This targeted approach could improve insulin sensitivity, reduce inflammation, and promote a healthier metabolic profile [199]. Additionally, FMT is being studied for its ability to restore microbial balance and improve neurological outcomes. FMT restores a healthy microbial community in the patient’s gut, which could have positive impacts on the GBA and neurological health [232234]. Therapeutically, there is potential for using appendix-derived microbiota in FMT for individuals with depression or anxiety [235]. This targeted approach could modulate the GBA and improve neurological health.

Engineered microbes and gene therapy

Potential live microbial biotherapeutics are made by engineered resident gut bacteria, which also make research easier into the effects of microorganisms and their metabolites on human health. The roles of the GM and its metabolome in the gut can be investigated in a more regulated, physiological setting by altering gut bacteria [236]. Different methods used to derive the desired engineered microbe include metabolic modifications and genetic modifications by loss of or gain of function [236]. For instance, In vitro investigations by the deletion of a bile salt hydrolase in B. thetaiotaomicron showed that decreased body weight and alterations to the host's metabolic, transcriptional profile, and immune response pathways are caused by the manipulation of tauro-β-muricholic bile acid [237]. In a similar study, CRISPR-Cas9-induced deletions were employed in Clostridium sporogenes to remove pathways that generate microbial metabolites. The impact of removing particular metabolites on host physiology was then examined. When genes for 10 C. sporogenes were knocked out, it produced microbial products IgA–modulatory activity [238]. The genes required for the whole manufacturing pathways known as lithocholic acid (LCA) and bile acids deoxycholic acid (DCA), which are known to inhibit the growth of Clostridium difficile and are two of the most prevalent small molecules in the microbiome of C. scindens, accumulate at approximately 500 μM. Subsequently, the genes required for the biosynthesis of LCA and DCA were refined and introduced into the genetically tractable producer C. sporogenes. [239].

Engineered gut bacterial were used in different studies to restore and rebalance GM [240242]. Irritable bowel syndrome and IBD are two GI disorders that have been treated with E. coli Nissle 1917 (EcN) [241, 242]. EcN is thought to prevent the formation of opportunistic infections by producing microcin proteins or siderophores that scavenge iron, which inhibits the growth of Salmonella spp. and other coliform enteropathogens [240, 242].

Developing a new class of engineered live bacterial therapeutics to treat human diseases is another promising advance of science [243]. To convert the comparatively innocuous substance 5-fluorocytosine (5-FC) into the cytotoxic compound 5-FU in situ, for instance, strains of Bifidobacterium have been modified to express cytosine deaminase [244]. In mice, co-administration of 5-FC with a B. infantis strain that produces cytosine deaminase-e dramatically suppressed tumor growth [245]. Similarly, to inhibit tumor growth in a mouse model, Li et al. built an EcN model to create cytotoxic chemicals, such as glidobactin, luminmide, and colibactin [246]. Targeting specific cancer cells has been demonstrated by several bacterial toxins. Targeted gene therapy against colon cancer cell lines overexpressing claudin-3 and/or − 4 can be achieved by using a translationally optimized Clostridium perfringens enterotoxin expression vector (optCPE), which enables the quick and efficient removal of tumor cells in both in vivo and in vitro [247].

Phage therapy

Phage therapy is an excellent strategy to treat infections. It has received notable attention for the effective eradication of multidrug-resistance bacteria and treating diseases [248]. By modularly switching phage tail components, a generalizable and effective technique for phage genome engineering was developed to reroute E. coli phage scaffolds to target pathogenic Yersinia and Klebsiella bacteria, and vice versa. The new target microorganisms were effectively eliminated by the synthetic phages [249]. According to a recent study, the lytic five-phage combination reduces inflammation and the severity of the disease by targeting susceptible and resistant members of the K. pneumoniae (Kp) clade, which is linked to IBD [248, 250]. Multidrug-resistant uropathogenic E. coli (UPEC) represents a significant global health system danger [251, 252]. According to the results of the phage-host interaction, the phage vB_Ec_ZCEC14 of the Straboviridae family has potent and consistent antibacterial activity against antibiotic-resistant UPEC at lower doses (MOI 0.1) [253]. In a 28-day double-blind, placebo-controlled crossover experiment, people ranging in weight from normal to overweight took bacteriophages. Phage eating was shown to be associated with decreases in fecal E. coli levels. Additionally, a lower percentage of taxa closest related to Clostridium perfringens and an increase in members of the butyrate-producing genus Eubacterium were noted [254]. For a duration of 28 days, healthy persons who self-reported experiencing GI distress were included in another study. They were instructed to take one 15-mg capsule containing four different strains of bacteriophages (LH01-Myoviridae, LL5-Siphoviridae, T4D-Myoviridae, and LL12-Myoviridae) or a placebo. While receiving the therapy and the placebo, participants also reported notable improvements in a number of GI discomfort symptoms [255]. Fusobacterium nucleatum (Fn) preferentially generates immunosuppressive myeloid-derived suppressor cells (MDSCs) to impede the host's anticancer immune response. This process is how Fn contributes to CRC. Using silver nanoparticles (AgNP) together with an M13 phage that selectively binds Fn demonstrated exceptional efficiency against Fn. According to research conducted in vivo and in vitro, M13@Ag therapy can scavenge Fn in the gut and reduce MDSC proliferation at the tumor site [256]. Using a lytic phage cocktail showed potential for targeting Klebsiella pneumoniae and Enterococcus gallinarum to treat primary sclerosing cholangitis [257].

Conclusion and future prospects

The AM emerges as a crucial contributor to human health, challenging conventional perceptions. It serves as a sanctuary for beneficial bacteria, crucial for gut health and immune system modulation. While direct evidence of the AM’s role in different diseases is still developing, the concept of using it as a therapeutic reservoir is promising and warrants further investigation. Research revealed that autoimmunity might be induced by FMT when transferred from autoimmune patients to healthy mice [89]. It is assumed that healthy subjects contain a balanced GM. When a transferred microbe by FTM causes autoimmune disease in healthy subjects, it means the transferred microbes might become dominant over the commensal flora. Issues like this could open a new era of research. Moreover, there are conflicting outcomes in research when working on the role of GM in some disease progression like PD. The conflicting evidence on the relationship between appendectomy and PD underscores the complexity of the disease and the need for a nuanced understanding of the GBA. Therefore, clinicians should consider appendectomy history when assessing PD, but more studies are required to clarify this relationship and explore therapeutic possibilities. Postbiotics could signify an advanced therapeutic approach if the microbial metabolites that are part of them can work synergistically on the target site (like the gut-vascular axis). In this instance, the intravenously supplied metabolites of bacteria might enter the bloodstream and provide direct action on, which is a novel indirect upgrade on vascular function brought about by the restoration of the intestinal microbiota’s homeostasis [211]. However, it is a promising advancement of science and could be applied to investigate other diseases as well.

Future research should focus on several key areas. There is a need for well-designed clinical trials and longitudinal studies to explore the role of AM in health and disease comprehensively. These studies should aim to understand the long-term effects of manipulating AM and its impact on various health conditions, including autoimmune diseases, neurodegenerative disorders, and metabolic syndromes. Research on gut fungi and parasites is significantly lacking compared to the gut bacteria. Future research should aim to elucidate the roles of gut fungi, parasites, and viruses, and their role in disease progression and their potential as therapeutic targets. Understanding the interactions between these microorganisms and the host immune system could lead to novel therapies for various diseases. The targeted change of microbiota by either restoringfunctions that are missing or eliminating functions which are harmful, may result in new methods to avoid or cure various ailments [21]. Future research promises innovative therapies harnessing the appendix's microbiota to enhance immune function and combat disease. Despite current limitations, particularly in human studies, understanding the appendix's complex symbiosis with the GM holds transformative potential for personalized medicine. As we unravel its complexities, the appendix represents the profound interplay between humans and their microbial counterparts, offering avenues for revolutionary healthcare advancements.

Acknowledgements

Open Access Funding was provided by the University of Applied Sciences Technikum Wien.

Abbreviations

AM

Appendix microbiome

AD

Auto-immune disease

ALS

Amyotrophic lateral sclerosis

ASD

Autism spectrum disorder

CD

Crohn’s disease

CNS

Central nervous system

CRC

Colorectal cancer

CVDs

Cardiovascular diseases

FMT

Fecal microbial transplantation

GBA

Gut-brain axis

GI

Gastrointestinal

GM

Gut microbiota

IBD

Inflammatory bowel disease

LDL

Low-density lipoprotein

LPS

Lipopolysaccharides

NDs

Neurological disorders

PD

Parkinson disease

PKC

Protein kinase C

SCFAs

Short-chain fatty acids

TLR

Toll-like receptor

UC

Ulcerative colitis

Author contributions

The authors’ responsibilities were as follows—MSS and TI: conceived the study and wrote the original draft; MSS, TI, NTT, MKIB, D, MAH, and ML: performed investigation and formal analysis; MSS, TI, D, MAH, and ML: reviewed and edited the manuscript; MAH and ML: supervised the study; All authors have read and agreed to the published version of the manuscript.

Funding

Not applicable.

Availability of data and materials

No datasets were generated or analysed during the current study.

Declarations

Competing interests

The authors declare no competing interests.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Md Shahjalal Sagor and Tarequl Islam have contributed equally to this work.

Contributor Information

Danishuddin, Email: danish23@yu.ac.kr.

Md Azizul Haque, Email: azizul@ynu.ac.kr.

Maximilian Lackner, Email: maximilian.lackner@technikum-wien.at.

References

  • 1.Girard-Madoux MJH, Gomez de Agüero M, Ganal-Vonarburg SC, Mooser C, Belz GT, Macpherson AJ, et al. The immunological functions of the appendix: an example of redundancy? Semin Immunol. 2018;36:31–44. 10.1016/j.smim.2018.02.005. [DOI] [PubMed] [Google Scholar]
  • 2.Petruzziello C, Saviano A, Ojetti V. Probiotics, the immune response and acute appendicitis: a review. Vaccines (Basel). 2023;11:1170. 10.3390/VACCINES11071170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Dictionary OL. Oxford latin dictionary. Oxford: Clarendon Press; 1968. [Google Scholar]
  • 4.Walker-Esbaugh C, McCarthy LH, Sparks RA, Dunmore CW. Dunmore and Fleischer’s medical terminology: exercises in etymology. 3rd ed. Philadelphia: FA Davis Co.; 2004. [Google Scholar]
  • 5.Dorland WAN. Dorland’s illustrated medical dictionary. 33rd ed. Amsterdam: Elsevier; 2020. [Google Scholar]
  • 6.Lee MS, Sulit A, Frizelle F, Purcell R. The microbiome in adult acute appendicitis. Gut Microbiome. 2022;3:e8. 10.1017/GMB.2022.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Cai S, Fan Y, Zhang B, Lin J, Yang X, Liu Y, et al. Appendectomy is associated with alteration of human gut bacterial and fungal communities. Front Microbiol. 2021;12:724980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Smith HF, Parker W, Kotzé SH, Laurin M. Multiple independent appearances of the cecal appendix in mammalian evolution and an investigation of related ecological and anatomical factors. C R Palevol. 2013;12:339–54. 10.1016/J.CRPV.2012.12.001. [Google Scholar]
  • 9.Smith HF, Heather Smith CF. A review of the function and evolution of the cecal appendix. Anat Rec. 2023;306:972–82. 10.1002/AR.24917. [DOI] [PubMed] [Google Scholar]
  • 10.Smith HF, Parker W, Kotzé SH, Laurin M. Morphological evolution of the mammalian cecum and cecal appendix. C R Palevol. 2017;16:39–57. 10.1016/J.CRPV.2016.06.001. [Google Scholar]
  • 11.Laurin M, Lou EM, Parker W. The cecal appendix: one more immune component with a function disturbed by post-industrial culture. Anat Rec Adv Integr Anat Evol Biol. 2011;294:567–79. [DOI] [PubMed] [Google Scholar]
  • 12.Collard MK, Bardin J, Marquet B, Laurin M, Ogier-Denis É. Correlation between the presence of a cecal appendix and reduced diarrhea severity in primates: new insights into the presumed function of the appendix. Sci Rep. 2023;13:1–10. 10.1038/s41598-023-43070-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Smith HF, Fisher RE, Everett ML, Thomas AD, Randal Bollinger R, Parker W. Comparative anatomy and phylogenetic distribution of the mammalian cecal appendix. J Evol Biol. 2009;22:1984–99. 10.1111/J.1420-9101.2009.01809.X. [DOI] [PubMed] [Google Scholar]
  • 14.Bininda-Emonds ORP, Cardillo M, Jones KE, MacPhee RDE, Beck RMD, Grenyer R, et al. The delayed rise of present-day mammals. Nature. 2007;446:507–12. 10.1038/nature05634. [DOI] [PubMed] [Google Scholar]
  • 15.Vaher K, Bogaert D, Richardson H, Boardman JP. Microbiome-gut-brain axis in brain development, cognition and behavior during infancy and early childhood. Dev Rev. 2022;66:101038. [Google Scholar]
  • 16.Lahiri D, Nag M, Dey A, Sayyed RZ, Ray RR. Interactions of microbiome for gut-brain axis health. In: Sayyed RZ, Khan M, editors. Microbiome-gut-brain axis: implications on health. Singapore: Springer; 2022. p. 139–51. [Google Scholar]
  • 17.Wang X, Sun X, Chu J, Sun W, Yan S, Wang Y. Gut microbiota and microbiota-derived metabolites in colorectal cancer: enemy or friend. World J Microbiol Biotechnol. 2023;39:291. [DOI] [PubMed] [Google Scholar]
  • 18.Liang C-S, Bai Y-M, Hsu J-W, Huang K-L, Chu C-S, Yeh T-C, et al. The risk of Alzheimer’s disease after acute appendicitis with or without appendectomy. J Am Med Dir Assoc. 2022;23:601–7. [DOI] [PubMed] [Google Scholar]
  • 19.Ooi H, Viswanathan S, Botross N, Eow G, Chin Y, Kanesalingam R, et al. Association between appendectomy and demyelinating disorders in subjects 40 years and older. Neurol India. 2020;68:850–5. 10.4103/0028-3886.293469. [DOI] [PubMed] [Google Scholar]
  • 20.Zaid M, Saleemi F, Usama RM, Tahir A, Azhar A, Eljack MMF, et al. Neuromyelitis optica spectrum disorder after post appendectomy peritonitis: a case report. Radiol Case Rep. 2024;19:6648–51. 10.1016/J.RADCR.2024.09.078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Young VB. The role of the microbiome in human health and disease: an introduction for clinicians. BMJ. 2017. 10.1136/BMJ.J831. [DOI] [PubMed] [Google Scholar]
  • 22.Arneth BM. Gut–brain axis biochemical signalling from the gastrointestinal tract to the central nervous system: gut dysbiosis and altered brain function. Postgrad Med J. 2018;94:446–52. [DOI] [PubMed] [Google Scholar]
  • 23.Chakrabarti A, Geurts L, Hoyles L, Iozzo P, Kraneveld AD, La Fata G, et al. The microbiota–gut–brain axis: pathways to better brain health. Perspectives on what we know, what we need to investigate and how to put knowledge into practice. Cell Mol Life Sci. 2022;79:80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Gómez-Duarte OG, Ogra PL. Editorial: impact of microbiome on gut mucosal immunity in health and disease. Front Immunol. 2022;13:1005156. 10.3389/FIMMU.2022.1005156/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zyoud SH, Smale S, Waring WS, Sweileh WM, Al-Jabi SW. Global research trends in microbiome-gut-brain axis during 2009–2018: a bibliometric and visualized study. BMC Gastroenterol. 2019;19:1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Kirkik D, Tas SK. Unveiling the intricacies of irritable bowel syndrome. World J Gastroenterol. 2024;30:4763. 10.3748/WJG.V30.I44.4763. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Schroeder BO, Bäckhed F. Signals from the gut microbiota to distant organs in physiology and disease. Nat Med. 2016;22:1079–89. 10.1038/NM.4185. [DOI] [PubMed] [Google Scholar]
  • 28.Granados-Martinez C, Alfageme-Lopez N, Navarro-Oviedo M, Nieto-Vaquero C, Cuartero MI, Diaz-Benito B, et al. Gut microbiota, bacterial translocation, and stroke: current knowledge and future directions. Biomedicines. 2024;12:2781. 10.3390/BIOMEDICINES12122781. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Bashir Y, Khan AU. The interplay between the gut-brain axis and the microbiome: a perspective on psychiatric and neurodegenerative disorders. Front Neurosci. 2022;16:1030694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Santos J, Maran PL, Rodríguez-Urrutia A. Stress, microbiota, and the gut–brain axis in mental and digestive health. Med Clin (Barc). 2025. 10.1016/J.MEDCLI.2024.11.023. [DOI] [PubMed] [Google Scholar]
  • 31.LaGreca M, Hutchinson DR, Skehan L. The microbiome and neurotransmitter activity. J Sci Med Art. 2021. 10.37714/josam.v3i2.90. [Google Scholar]
  • 32.Hook DW, Porter SJ, Herzog C. Dimensions: building context for search and evaluation. Front Res Metr Anal. 2018;3:386181. 10.3389/FRMA.2018.00023/BIBTEX. [Google Scholar]
  • 33.Moher D, Liberati A, Tetzlaff J, Altman DG, Antes G, Atkins D, et al. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med. 2009. 10.1371/JOURNAL.PMED.1000097. [PMC free article] [PubMed] [Google Scholar]
  • 34.van Eck NJ, Waltman L. Software survey: VOSviewer, a computer program for bibliometric mapping. Scientometrics. 2010;84:523–38. 10.1007/S11192-009-0146-3/FIGURES/7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Cully M. Antibiotics alter the gut microbiome and host health. Nat Res. 2019;15:s19. [Google Scholar]
  • 36.Asnicar F, Berry SE, Valdes AM, Nguyen LH, Piccinno G, Drew DA, et al. Microbiome connections with host metabolism and habitual diet from 1,098 deeply phenotyped individuals. Nat Med. 2021;27:321–32. 10.1038/S41591-020-01183-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Deleemans JM, Chleilat F, Reimer RA, Henning JW, Baydoun M, Piedalue KA, et al. The chemo-gut study: Investigating the long-term effects of chemotherapy on gut microbiota, metabolic, immune, psychological and cognitive parameters in young adult Cancer survivors; Study protocol. BMC Cancer. 2019;19:1–11. 10.1186/S12885-019-6473-8/TABLES/1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020. 10.1038/s41422-020-0332-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.He P, Yu L, Tian F, Zhang H, Chen W, Zhai Q. Dietary patterns and gut microbiota: the crucial actors in inflammatory bowel disease. Adv Nutr. 2022;13:1628–51. 10.1093/ADVANCES/NMAC029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Pendyala S, Walker JM, Holt PR. A high-fat diet is associated with endotoxemia that originates from the gut. Gastroenterology. 2012. 10.1053/J.GASTRO.2012.01.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Turnbaugh PJ, Ridaura VK, Faith JJ, Rey FE, Knight R, Gordon JI. The effect of diet on the human gut microbiome: a metagenomic analysis in humanized gnotobiotic mice. Sci Transl Med. 2009;1:6ra14. 10.1126/SCITRANSLMED.3000322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Berg AM, Kelly CP, Farraye FA. Clostridium difficile infection in the inflammatory bowel disease patient. Inflamm Bowel Dis. 2013;19:194–204. 10.1002/IBD.22964. [DOI] [PubMed] [Google Scholar]
  • 43.Begley M, Hill C, Gahan CGM. Bile salt hydrolase activity in probiotics. Appl Environ Microbiol. 2006;72:1729. 10.1128/AEM.72.3.1729-1738.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Tong XN, Wang XZ, He XJ, Wang Z, Li WX. Effects of antibiotics on microbial community structure and microbial functions in constructed wetlands treated with artificial root exudates. Environ Sci Process Impacts. 2020;22:217–26. 10.1039/C9EM00458K. [DOI] [PubMed] [Google Scholar]
  • 45.Brown K, DeCoffe D, Molcan E, Gibson DL. Diet-induced dysbiosis of the intestinal microbiota and the effects on immunity and disease. Nutrients. 2012;4:1095. 10.3390/NU4081095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Zhao W, Hu Y, Li C, Li N, Zhu S, Tan X, et al. Transplantation of fecal microbiota from patients with alcoholism induces anxiety/depression behaviors and decreases brain mGluR1/PKC ε levels in mouse. BioFactors. 2020;46:38–54. 10.1002/biof.1567. [DOI] [PubMed] [Google Scholar]
  • 47.Hossain KS, Amarasena S, Mayengbam S. B vitamins and their roles in gut health. Microorganisms. 2022;10:1168. 10.3390/MICROORGANISMS10061168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kelly CJ, Alexeev EE, Farb L, Vickery TW, Zheng L, Eric LC, et al. Oral vitamin B12 supplement is delivered to the distal gut, altering the corrinoid profile and selectively depleting Bacteroides in C57BL/6 mice. Gut Microbes. 2019;10:654. 10.1080/19490976.2019.1597667. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Mehra A, Arora G, Sahni G, Kaur M, Singh H, Singh B, et al. Gut microbiota and autism spectrum disorder: from pathogenesis to potential therapeutic perspectives. J Tradit Complement Med. 2022;13:135. 10.1016/J.JTCME.2022.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Xie G, Zhou Q, Qiu CZ, Dai WK, Wang HP, Li YH, et al. Ketogenic diet poses a significant effect on imbalanced gut microbiota in infants with refractory epilepsy. World J Gastroenterol. 2017;23:6164–71. 10.3748/WJG.V23.I33.6164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Lee S, Jang EJ, Jo J, Park SJ, Ryu HG. Long-term impacts of appendectomy associated with increased incidence of inflammatory bowel disease, infection, and colorectal cancer. Int J Colorectal Dis. 2021;36:1643–52. 10.1007/S00384-021-03886-X/METRICS. [DOI] [PubMed] [Google Scholar]
  • 52.Sugihara K, Kamada N. Metabolic network of the gut microbiota in inflammatory bowel disease. Inflamm Regen. 2024;44:1–11. 10.1186/S41232-024-00321-W/FIGURES/3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Geboes K. Appendiceal function and dysfunction: what are the implications for inflammatory bowel disease? Nat Clin Pract Gastroenterol Hepatol. 2005;2:338–9. 10.1038/ncpgasthep0238. [DOI] [PubMed] [Google Scholar]
  • 54.Islam T, Saha O, Hridoy L, Hasan M, Sultana S, Marzan S, et al. Comparison between reduced susceptibility to disinfectants and multidrug resistance among hospital isolates of Pseudomonas aeruginosa and Staphylococcus aureus in Bangladesh. Bagcilar Med Bull. 2017;2:88–97. 10.5350/BMB20170828103017. [Google Scholar]
  • 55.Sagor MS, Hossain MS, Islam T, Mahmud MA, Miah MS, Karim MR, et al. Phenotypic and genotypic antibiotic resistance and virulence profiling of enterococcus faecalis isolated from poultry at two major districts in Bangladesh. Pak Vet J. 2022;42:153–60. 10.29261/pakvetj/2022.019. [Google Scholar]
  • 56.Islam T, Kubra K, Hassan Chowdhury MM. Prevalence of methicillin-resistant Staphylococcus aureus in hospitals in Chittagong, Bangladesh: a threat of nosocomial infection. J Microsc Ultrastruct. 2018;6:188–91. 10.4103/JMAU.JMAU_33_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Debnath T, Bhowmik S, Islam T, Hassan CM. Presence of multidrug-resistant bacteria on mobile phones of healthcare workers accelerates the spread of nosocomial infection and regarded as a threat to public health in Bangladesh. J Microsc Ultrastruct. 2018;6:165. 10.4103/JMAU.JMAU_30_18. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Islam T, Haque MA, Barai HR, Istiaq A, Kim JJ. Antibiotic resistance in plant pathogenic bacteria: recent data and environmental impact of unchecked use and the potential of biocontrol agents as an eco-friendly alternative. Plants. 2024;13:1135. 10.3390/PLANTS13081135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Patangia DV, Anthony Ryan C, Dempsey E, Paul Ross R, Stanton C. Impact of antibiotics on the human microbiome and consequences for host health. Microbiologyopen. 2022;11:e1260. 10.1002/MBO3.1260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Brody H. The gut microbiome. Nature. 2020;577:S5. 10.1038/D41586-020-00194-2. [DOI] [PubMed] [Google Scholar]
  • 61.Emanuele E, Orsi P, Boso M, Broglia D, Brondino N, Barale F, et al. Low-grade endotoxemia in patients with severe autism. Neurosci Lett. 2010;471:162–5. 10.1016/J.NEULET.2010.01.033. [DOI] [PubMed] [Google Scholar]
  • 62.Islam T, Tamanna NT, Sagor MS, Zaki RM, Rabbee MF, Lackner M. Antimicrobial peptides: a promising solution to the rising threat of antibiotic resistance. Pharmaceutics. 2024;16:1542. 10.3390/PHARMACEUTICS16121542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Kirsten TB, Chaves-Kirsten GP, Chaible LM, Silva AC, Martins DO, Britto LRG, et al. Hypoactivity of the central dopaminergic system and autistic-like behavior induced by a single early prenatal exposure to lipopolysaccharide. J Neurosci Res. 2012;90:1903–12. 10.1002/JNR.23089. [DOI] [PubMed] [Google Scholar]
  • 64.Ervin SM, Ramanan SV, Bhatt AP. Relationship between the gut microbiome and systemic chemotherapy. Dig Dis Sci. 2020;65:874–84. 10.1007/S10620-020-06119-3/METRICS. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Huang X, Fang Q, Rao T, Zhou L, Zeng X, Tan Z, et al. Leucovorin ameliorated methotrexate induced intestinal toxicity via modulation of the gut microbiota. Toxicol Appl Pharmacol. 2020. 10.1016/J.TAAP.2020.114900. [DOI] [PubMed] [Google Scholar]
  • 66.Sougiannis AT, VanderVeen BN, Enos RT, Velazquez KT, Bader JE, Carson M, et al. Impact of 5 fluorouracil chemotherapy on gut inflammation, functional parameters, and gut microbiota. Brain Behav Immun. 2019;80:44–55. 10.1016/J.BBI.2019.02.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Ying M, Yu Q, Zheng B, Wang H, Wang J, Chen S, et al. Cultured Cordyceps sinensis polysaccharides modulate intestinal mucosal immunity and gut microbiota in cyclophosphamide-treated mice. Carbohydr Polym. 2020. 10.1016/J.CARBPOL.2020.115957. [DOI] [PubMed] [Google Scholar]
  • 68.Zhang X, di Chen B, Zhao L, dan, Li H. The gut microbiota: emerging evidence in autoimmune diseases. Trends Mol Med. 2020;26:862–73. 10.1016/J.MOLMED.2020.04.001. [DOI] [PubMed] [Google Scholar]
  • 69.Panebianco C, Adamberg K, Jaagura M, Copetti M, Fontana A, Adamberg S, et al. Influence of gemcitabine chemotherapy on the microbiota of pancreatic cancer xenografted mice. Cancer Chemother Pharmacol. 2018;81:773–82. 10.1007/S00280-018-3549-0. [DOI] [PubMed] [Google Scholar]
  • 70.Youssef O, Lahti L, Kokkola A, Karla T, Tikkanen M, Ehsan H, et al. Stool microbiota composition differs in patients with stomach, colon, and rectal neoplasms. Dig Dis Sci. 2018;63:2950–8. 10.1007/S10620-018-5190-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Deng X, Li Z, Li G, Li B, Jin X, Lyu G. Comparison of microbiota in patients treated by surgery or chemotherapy by 16S rRNA sequencing reveals potential biomarkers for colorectal cancer therapy. Front Microbiol. 2018. 10.3389/FMICB.2018.01607. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Wei L, Wen X-S, Xian CJ. Chemotherapy-induced intestinal microbiota dysbiosis impairs mucosal homeostasis by modulating toll-like receptor signaling pathways. Int J Mol Sci. 2021;22:9474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Li W, Liu J, Tan H, Yang C, Ren L, Liu Q, et al. Genetic effects on the gut microbiota assemblages of hybrid fish from parents with different feeding habits. Front Microbiol. 2018;9:419640. 10.3389/FMICB.2018.02972/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Muhammad AY, Amonov M, Baig AA, Alvi FJ. Gut microbiome: an intersection between human genome, diet, and epigenetics. Adv Gut Microbiome Res. 2024;2024:6707728. 10.1155/2024/6707728. [Google Scholar]
  • 75.Qin Y, Havulinna AS, Liu Y, Jousilahti P, Ritchie SC, Tokolyi A, et al. Combined effects of host genetics and diet on human gut microbiota and incident disease in a single population cohort. Nat Genet. 2022;54:134–42. 10.1038/s41588-021-00991-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chambers JC, Zhang W, Sehmi JS, Li X, Wass MN, Van der Harst P, et al. Genome-wide association study identifies loci influencing concentrations of liver enzymes in plasma. Nat Genet. 2011;43:1131–8. 10.1038/NG.970. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Kobayashi KS, Chamaillard M, Ogura Y, Henegariu O, Inohara N, Nuñez G, et al. Nod2-dependent regulation of innate and adaptive immunity in the intestinal tract. Science. 1979;2005(307):731–4. 10.1126/SCIENCE.1104911. [DOI] [PubMed] [Google Scholar]
  • 78.Goodrich JK, Davenport ER, Clark AG, Ley RE. The relationship between the human genome and microbiome comes into view. Annu Rev Genet. 2017;51:413–33. 10.1146/ANNUREV-GENET-110711-155532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Gubatan J, Boye TL, Temby M, Sojwal RS, Holman DR, Sinha SR, et al. Gut microbiome in inflammatory bowel disease: role in pathogenesis, dietary modulation, and colitis-associated colon cancer. Microorganisms. 2022;10:1371. 10.3390/MICROORGANISMS10071371. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Sadeghpour HF. Gut microbiota and autoimmune diseases: mechanisms, treatment, challenges, and future recommendations. Curr Clin Microbiol Rep. 2024;11:18–33. 10.1007/S40588-023-00213-6/TABLES/2. [Google Scholar]
  • 81.Yu CH, Chang CN, Wang CC. Causative microbes and antibiotic susceptibility of acute appendicitis in adults and children. Pediatr Neonatol. 2024;65:159–64. 10.1016/j.pedneo.2023.08.003. [DOI] [PubMed] [Google Scholar]
  • 82.Cimpean S, Barranquero AG, Surdeanu I, Cadiere B, Cadiere G-B. Implications of bacteriological study in complicated and uncomplicated acute appendicitis. Ann Coloproctol. 2022. 10.3393/AC.2022.00157.0022. [DOI] [PubMed] [Google Scholar]
  • 83.Blohs M, Mahnert A, Brunnader K, Flucher C, Castellani C, Till H, et al. Acute appendicitis manifests as two microbiome state types with oral pathogens influencing severity. Gut Microbes. 2023. 10.1080/19490976.2022.2145845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Rogers MB, Brower-Sinning R, Firek B, Zhong D, Morowitz MJ. Acute appendicitis in children is associated with a local expansion of fusobacteria. Clin Infect Dis. 2016;63:71–8. 10.1093/CID/CIW208. [DOI] [PubMed] [Google Scholar]
  • 85.Aiyoshi T, Kakihara T, Watanabe E, Tanaka N, Ogata Y, Masuoka H, et al. A comprehensive microbial analysis of pediatric patients with acute appendicitis. J Microbiol Immunol Infect. 2023;56:695–704. 10.1016/J.JMII.2023.03.006. [DOI] [PubMed] [Google Scholar]
  • 86.Oh SJ, Pimentel MM, Leite GGS, Celly S, Villanueva-Millan MJ, Lacsina I, et al. Acute appendicitis is associated with appendiceal microbiome changes including elevated Campylobacter jejuni levels. BMJ Open Gastroenterol. 2020. 10.1136/bmjgast-2020-000412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ahmed J, Reddy BS, Mølbak L, Leser TD, MacFie J. Impact of probiotics on colonic microflora in patients with colitis: a prospective double blind randomised crossover study. Int J Surg. 2013;11:1131–6. [DOI] [PubMed] [Google Scholar]
  • 88.Arjomand Fard N, Armstrong H, Perry T, Wine E. Appendix and ulcerative colitis: a key to explaining the pathogenesis and directing novel therapies? Inflamm Bowel Dis. 2023;29:151–60. 10.1093/IBD/IZAC106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ye Z, Zhang N, Wu C, Zhang X, Wang Q, Huang X, et al. A metagenomic study of the gut microbiome in Behcet’s disease. Microbiome. 2018. 10.1186/S40168-018-0520-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Kirkik D, Kalkanli Tas S, Tanoglu A. Unraveling the blood microbiome: novel insights into inflammasome responses in Crohn’s disease. Eur J Gastroenterol Hepatol. 2024;36:975–84. 10.1097/MEG.0000000000002695. [DOI] [PubMed] [Google Scholar]
  • 91.Clooney AG, Sutton TDS, Shkoporov AN, Holohan RK, Daly KM, O’Regan O, et al. Whole-virome analysis sheds light on viral dark matter in inflammatory bowel disease. Cell Host Microbe. 2019;26:764-778.e5. 10.1016/J.CHOM.2019.10.009. [DOI] [PubMed] [Google Scholar]
  • 92.Guzzo GL, Mittinty MN, Llamas B, Andrews JM, Weyrich LS. Individuals with inflammatory bowel disease have an altered gut microbiome composition of fungi and protozoa. Microorganisms. 2022;10:1910. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Xu H, Liu M, Cao J, Li X, Fan D, Xia Y, et al. The dynamic interplay between the gut microbiota and autoimmune diseases. J Immunol Res. 2019;2019:7546047. 10.1155/2019/7546047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Rothhammer V, Quintana FJ. The aryl hydrocarbon receptor: an environmental sensor integrating immune responses in health and disease. Nat Rev Immunol. 2019;19:184–97. 10.1038/S41577-019-0125-8. [DOI] [PubMed] [Google Scholar]
  • 95.Kawajiri K, Fujii-Kuriyama Y. The aryl hydrocarbon receptor: a multifunctional chemical sensor for host defense and homeostatic maintenance. Exp Anim. 2017;66:75–89. 10.1538/EXPANIM.16-0092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Qiu J, Guo X, Chen Z, Ming E, He L, Sonnenberg GF, Artis D, et al. Group 3 innate lymphoid cells inhibit T-cell-mediated intestinal inflammation through aryl hydrocarbon receptor signaling and regulation of microflora. Immunity. 2013;39:386–99. 10.1016/J.IMMUNI.2013.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Stedtfeld RD, Chai B, Crawford RB, Stedtfeld TM, Williams MR, Xiangwen S, et al. Modulatory influence of segmented filamentous bacteria on transcriptomic response of gnotobiotic mice exposed to tcdd. Front Microbiol. 2017;8:286560. 10.3389/FMICB.2017.01708/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Dehner C, Fine R, Kriegel MA. The microbiome in systemic autoimmune disease: mechanistic insights from recent studies. Curr Opin Rheumatol. 2019;31:201–7. 10.1097/BOR.0000000000000574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.López P, De Paz B, Rodríguez-Carrio J, Hevia A, Sánchez B, Margolles A, et al. Th17 responses and natural IgM antibodies are related to gut microbiota composition in systemic lupus erythematosus patients. Sci Rep. 2016. 10.1038/SREP24072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Azzouz D, Omarbekova A, Heguy A, Schwudke D, Gisch N, Rovin BH, et al. Lupus nephritis is linked to disease-activity associated expansions and immunity to a gut commensal. Ann Rheum Dis. 2019;78:947–56. 10.1136/ANNRHEUMDIS-2018-214856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Chen J, Wright K, Davis JM, Jeraldo P, Marietta EV, Murray J, et al. An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis. Genome Med. 2016;8:1–14. 10.1186/S13073-016-0299-7/FIGURES/6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Chiang HI, Li JR, Liu CC, Liu PY, Chen HH, Chen YM, et al. An association of gut microbiota with different phenotypes in chinese patients with rheumatoid arthritis. J Clin Med. 2019. 10.3390/JCM8111770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski C, et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife. 2013. 10.7554/ELIFE.01202.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Yap DRY, Lui RN, Samol J, Ngeow J, Sung JJYY, Wong SH. Beyond a vestigial organ: effects of the appendix on gut microbiome and colorectal cancer. J Gastroenterol Hepatol. 2024;39:826–35. 10.1111/JGH.16497. [DOI] [PubMed] [Google Scholar]
  • 105.McKnight DT, Huerlimann R, Bower DS, Schwarzkopf L, Alford RA, Zenger KR. The interplay of fungal and bacterial microbiomes on rainforest frogs following a disease outbreak. Ecosphere. 2022;13: e4037. [Google Scholar]
  • 106.Viswanathan S, Parida S, Lingipilli BT, Krishnan R, Podipireddy DR, Muniraj N. Role of gut microbiota in breast cancer and drug resistance. Pathogens. 2023;12:468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Sampsell K, Hao D, Reimer RA. The gut microbiota: a potential gateway to improved health outcomes in breast cancer treatment and survivorship. Int J Mol Sci. 2020;21:9239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Sánchez-Alcoholado L, Ramos-Molina B, Otero A, Laborda-Illanes A, Ordóñez R, Medina JA, et al. The role of the gut microbiome in colorectal cancer development and therapy response. Cancers (Basel). 2020;12:1406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Dalal N, Jalandra R, Bayal N, Yadav AK, Harshulika N, Sharma M, et al. Gut microbiota-derived metabolites in CRC progression and causation. J Cancer Res Clin Oncol. 2021;147:3141–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Long Y, Tang L, Zhou Y, Zhao S, Zhu H. Causal relationship between gut microbiota and cancers: a two-sample Mendelian randomisation study. BMC Med. 2023;21:66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Su Q, Jin C, Bo Z, Yang Y, Wang J, Wang J, et al. Association between gut microbiota and gastrointestinal cancer: a two-sample bi-directional Mendelian randomization study. Front Microbiol. 2023;14:1181328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Vietsch EE, Latifi D, Verheij M, van der Oost EWA, de Wilde RF, Haen R, et al. B cell immune profiles in dysbiotic vermiform appendixes of pancreatic cancer patients. Front Immunol. 2023. 10.3389/FIMMU.2023.1230306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Ajami NJ, Wargo JA. AI finds microbial signatures in tumours and blood across cancer types. Nature. 2020;579:502–3. [DOI] [PubMed] [Google Scholar]
  • 114.Yang X, An H, He Y, Fu G, Jiang Z. Comprehensive analysis of microbiota signature across 32 cancer types. Front Oncol. 2023;13:1127225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Zhao LY, Mei JX, Yu G, Lei L, Zhang WH, Liu K, et al. Role of the gut microbiota in anticancer therapy: from molecular mechanisms to clinical applications. Signal Transduct Target Ther. 2023;8:1–27. 10.1038/s41392-023-01406-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Tortora SC, Bodiwala VM, Quinn A, Martello LA, Vignesh S. Microbiome and colorectal carcinogenesis: linked mechanisms and racial differences. World J Gastrointest Oncol. 2022;14:375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Sadrekarimi H, Gardanova ZR, Bakhshesh M, Ebrahimzadeh F, Yaseri AF, Thangavelu L, et al. Emerging role of human microbiome in cancer development and response to therapy: special focus on intestinal microflora. J Transl Med. 2022. 10.1186/S12967-022-03492-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Nougayrède J-P, Homburg S, Taieb F, Boury M, Brzuszkiewicz E, Gottschalk G, et al. Escherichia coli induces DNA double-strand breaks in eukaryotic cells. Science. 1979;2006(313):848–51. [DOI] [PubMed] [Google Scholar]
  • 119.Álvarez-Mercado AI, del Valle CA, Fernández MF, Fontana L. Gut microbiota and breast cancer: the dual role of microbes. Cancers (Basel). 2023;15:443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wroblewski LE, Peek RM Jr, Wilson KT. Helicobacter pylori and gastric cancer: factors that modulate disease risk. Clin Microbiol Rev. 2010;23:713–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Konturek SJ, Starzynska T, Konturek PC, Karczewska E, Marlicz K, Lawniczak M, et al. Helicobacter pylori and CagA status, serum gastrin, interleukin-8 and gastric acid secretion in gastric cancer. Scand J Gastroenterol. 2002;37:891–8. [DOI] [PubMed] [Google Scholar]
  • 122.Löwenmark T, Löfgren-Burström A, Zingmark C, Ljuslinder I, Dahlberg M, Edin S, et al. Tumour colonisation of Parvimonas micra is associated with decreased survival in colorectal cancer patients. Cancers (Basel). 2022;14:5937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Bergsten E, Mestivier D, Donnadieu F, Pedron T, Barau C, Meda LT, et al. Parvimonas micra, an oral pathobiont associated with colorectal cancer, epigenetically reprograms human colonocytes. Gut Microbes. 2023;15:2265138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Wang X, Allen TD, May RJ, Lightfoot S, Houchen CW, Huycke MM. Enterococcus faecalis induces aneuploidy and tetraploidy in colonic epithelial cells through a bystander effect. Cancer Res. 2008;68:9909–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Cheng WT, Kantilal HK, Davamani F. The mechanism of Bacteroides fragilis toxin contributes to colon cancer formation. Malays J Med Sci. 2020;27:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Zheng M, Han R, Yuan Y, Xing Y, Zhang W, Sun Z, et al. The role of Akkermansia muciniphila in inflammatory bowel disease: Current knowledge and perspectives. Front Immunol. 2023;13:1089600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Zong Z, Zhou F, Zhang L. The fungal mycobiome: a new hallmark of cancer revealed by pan-cancer analyses. Signal Transduct Target Ther. 2023;8:50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Kozlov M. Do fungi lurking inside cancers speed their growth? Nature. 2022. 10.1038/d41586-022-03074-z. [DOI] [PubMed] [Google Scholar]
  • 129.Camara A, Konate S, Tidjani Alou M, Kodio A, Togo AH, Cortaredona S, et al. Clinical evidence of the role of Methanobrevibacter smithii in severe acute malnutrition. Sci Rep. 2021;11:5426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Houshyar Y, Massimino L, Lamparelli LA, Danese S, Ungaro F. Going beyond bacteria: uncovering the role of archaeome and mycobiome in inflammatory bowel disease. Front Physiol. 2021;12:783295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Rebersek M. Gut microbiome and its role in colorectal cancer. BMC Cancer. 2021;21:1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Sarmiento-Andrade Y, Suárez R, Quintero B, Garrochamba K, Chapela SP. Gut microbiota and obesity: New insights. Front Nutr. 2022;9:1018212. 10.3389/FNUT.2022.1018212/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Wei M, Huang F, Zhao L, Zhang Y, Yang W, Wang S, et al. A dysregulated bile acid-gut microbiota axis contributes to obesity susceptibility. EBioMedicine. 2020. 10.1016/J.EBIOM.2020.102766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Miranda VPN, Dos Santos Amorim PR, Bastos RR, De Faria ER, De Castro Moreira ME, Do Franceschini SCC, et al. Abundance of gut microbiota, concentration of short-chain fatty acids, and inflammatory markers associated with elevated body fat, overweight, and obesity in female adolescents. Mediators Inflamm. 2019. 10.1155/2019/7346863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Del Chierico F, Manco M, Gardini S, Guarrasi V, Russo A, Bianchi M, et al. Fecal microbiota signatures of insulin resistance, inflammation, and metabolic syndrome in youth with obesity: a pilot study. Acta Diabetol. 2021;58:1009–22. 10.1007/S00592-020-01669-4. [DOI] [PubMed] [Google Scholar]
  • 136.Pai CS, Wang CY, Hung WW, Hung WC, Tsai HJ, Chang CC, et al. Interrelationship of gut microbiota, obesity, body composition and insulin resistance in asians with type 2 diabetes mellitus. J Pers Med. 2022. 10.3390/JPM12040617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57:1470–81. 10.2337/DB07-1403. [DOI] [PubMed] [Google Scholar]
  • 138.Mei H, Li N, Zhang Y, Zhang D, Peng A, Tan YF, et al. Gut microbiota diversity and overweight/obesity in infancy: results from a nested case-control study. Curr Med Sci. 2022;42:210–6. 10.1007/S11596-021-2476-1. [DOI] [PubMed] [Google Scholar]
  • 139.Caesar R, Reigstad CS, Bäckhed HK, Reinhardt C, Ketonen M, Lundén GÖ, et al. Gut-derived lipopolysaccharide augments adipose macrophage accumulation but is not essential for impaired glucose or insulin tolerance in mice. Gut. 2012;61:1701–7. 10.1136/GUTJNL-2011-301689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Teng F, Klinger CN, Felix KM, Bradley CP, Wu E, Tran NL, et al. Gut microbiota drive autoimmune arthritis by promoting differentiation and migration of Peyer’s patch T follicular helper cells. Immunity. 2016;44:875–88. 10.1016/J.IMMUNI.2016.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kitamura K, Shionoya H, Suzuki S, Fukai R, Uda S, Abe C, et al. Oral and intestinal bacterial substances associated with disease activities in patients with rheumatoid arthritis: a cross-sectional clinical study. J Immunol Res. 2022. 10.1155/2022/6839356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Maeda Y, Kurakawa T, Umemoto E, Motooka D, Ito Y, Gotoh K, et al. Dysbiosis contributes to arthritis development via activation of autoreactive T cells in the intestine. Arthritis Rheumatol. 2016;68:2646–61. 10.1002/ART.39783. [DOI] [PubMed] [Google Scholar]
  • 143.Zheng Y, Bonfili L, Wei T, Eleuteri AM. Understanding the gut-brain axis and its therapeutic implications for neurodegenerative disorders. Nutrients. 2023;15:4631. 10.3390/NU15214631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Soni A, Verma A, Gupta P. Microbiota–gut–brain axis and neurodegenerative disorder. Singapore: Springer; 2022. p. 27–46. [Google Scholar]
  • 145.Wu HJ, Wu E. The role of gut microbiota in immune homeostasis and autoimmunity. Gut Microbes. 2012;3:4. 10.4161/GMIC.19320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Ma Q, Xing C, Long W, Wang HY, Liu Q, Wang RF. Impact of microbiota on central nervous system and neurological diseases: the gut-brain axis. J Neuroinflamm. 2019. 10.1186/s12974-019-1434-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Gan Y, Chen Y, Zhong H, Liu Z, Geng J, Wang H, et al. Gut microbes in central nervous system development and related disorders. Front Immunol. 2024;14:1288256. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Minagar A, Jabbour R, Jabbour H. The human gut microbiota: a dynamic biologic factory. Berlin: Springer; 2023. p. 1–16. [Google Scholar]
  • 149.Günther C, Rothhammer V, Karow M, Neurath M, Winner B. The gut-brain axis in inflammatory bowel disease—current and future perspectives. Int J Mol Sci. 2021;22:8870. 10.3390/IJMS22168870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Socała K, Doboszewska U, Szopa A, Serefko A, Włodarczyk M, Zielińska A, et al. The role of microbiota-gut-brain axis in neuropsychiatric and neurological disorders. Pharmacol Res. 2021;172:105840. 10.1016/J.PHRS.2021.105840. [DOI] [PubMed] [Google Scholar]
  • 151.Li N, Wang Q, Wang Y, Sun A, Lin Y, Jin Y, et al. Fecal microbiota transplantation from chronic unpredictable mild stress mice donors affects anxiety-like and depression-like behavior in recipient mice via the gut microbiota-inflammation-brain axis. Stress. 2019;22:592–602. 10.1080/10253890.2019.1617267. [DOI] [PubMed] [Google Scholar]
  • 152.Sun L, Zhang H, Cao Y, Wang C, Zhao C, Wang H, et al. Fluoxetine ameliorates dysbiosis in a depression model induced by chronic unpredicted mild stress in mice. Int J Med Sci. 2019;16:1260–70. 10.7150/ijms.37322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Golofast B, Vales K. The connection between microbiome and schizophrenia. Neurosci Biobehav Rev. 2020;108:712–31. 10.1016/J.NEUBIOREV.2019.12.011. [DOI] [PubMed] [Google Scholar]
  • 154.Yuan X, Zhang P, Wang Y, Liu Y, Li X, Kumar BU, et al. Changes in metabolism and microbiota after 24-week risperidone treatment in drug naïve, normal weight patients with first episode schizophrenia. Schizophr Res. 2018;201:299–306. 10.1016/J.SCHRES.2018.05.017. [DOI] [PubMed] [Google Scholar]
  • 155.Li S, Zhuo M, Huang X, Huang Y, Zhou J, Xiong D, et al. Altered gut microbiota associated with symptom severity in schizophrenia. PeerJ. 2020;8:9574. 10.7717/PEERJ.9574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Nguyen TT, Kosciolek T, Maldonado Y, Daly RE, Martin AS, McDonald D, et al. Differences in gut microbiome composition between persons with chronic schizophrenia and healthy comparison subjects. Schizophr Res. 2019;204:23–9. 10.1016/J.SCHRES.2018.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Zhang X, Tang B, Guo J. Parkinson’s disease and gut microbiota: from clinical to mechanistic and therapeutic studies. Transl Neurodegener. 2023;12:1–30. 10.1186/S40035-023-00392-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Nishiwaki H, Ueyama J, Ito M, Hamaguchi T, Takimoto K, Maeda T, et al. Meta-analysis of shotgun sequencing of gut microbiota in Parkinson’s disease. Npj Parkinson’s Dis. 2024;10:1–11. 10.1038/s41531-024-00724-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Nishiwaki H, Ito M, Ishida T, Hamaguchi T, Maeda T, Kashihara K, et al. Meta-analysis of gut dysbiosis in Parkinson’s Disease. Mov Disord. 2020;35:1626–35. 10.1002/MDS.28119. [DOI] [PubMed] [Google Scholar]
  • 160.Romano S, Savva GM, Bedarf JR, Charles IG, Hildebrand F, Narbad A. Meta-analysis of the Parkinson’s disease gut microbiome suggests alterations linked to intestinal inflammation. NPJ Parkinsons Dis. 2021;7:1–13. 10.1038/s41531-021-00156-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Shen T, Yue Y, He T, Huang C, Qu B, Lv W, et al. The association between the gut microbiota and parkinson’s disease, a meta-analysis. Front Aging Neurosci. 2021. 10.3389/FNAGI.2021.636545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Johnson ME, Stringer A, Bobrovskaya L. Rotenone induces gastrointestinal pathology and microbiota alterations in a rat model of Parkinson’s disease. Neurotoxicology. 2018;65:174–85. 10.1016/J.NEURO.2018.02.013. [DOI] [PubMed] [Google Scholar]
  • 163.Park KW, Woo HT, Hwang YS, Lee SH, Chung SJ. Appendectomy and the risk of Parkinson’s disease: a Korean nationwide study. Mov Disord Clin Pract. 2024;11:704. 10.1002/MDC3.14031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Wang L, Cao W, Xi MH, Li Y. Appendectomy and the risk of neurodegenerative diseases: a two-sample Mendelian randomization study. Asian J Surg. 2024;47:673–4. 10.1016/J.ASJSUR.2023.09.170. [DOI] [PubMed] [Google Scholar]
  • 165.Jain S, Rosenbaum PR, Reiter JG, Hill AS, Wolk DA, Hashemi S, et al. Risk of Parkinson’s disease after anaesthesia and surgery. British J Anaesth. 2022;128:e268. 10.1016/J.BJA.2021.12.046. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Nakahara K, Nakane S, Ishii K, Ikeda T, Ando Y. Gut microbiota of Parkinson’s disease in an appendectomy cohort: a preliminary study. Sci Rep. 2023;13:1–10. 10.1038/s41598-023-29219-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Boddy SL, Giovannelli I, Sassani M, Cooper-Knock J, Snyder MP, Segal E, et al. The gut microbiome: a key player in the complexity of amyotrophic lateral sclerosis (ALS). BMC Med. 2021;19:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Brenner D, Hiergeist A, Adis C, Mayer B, Gessner A, Ludolph AC, et al. The fecal microbiome of ALS patients. Neurobiol Aging. 2018;61:132–7. 10.1016/J.NEUROBIOLAGING.2017.09.023. [DOI] [PubMed] [Google Scholar]
  • 169.Fulceri F, Morelli M, Santocchi E, Cena H, Del Bianco T, Narzisi A, et al. Gastrointestinal symptoms and behavioral problems in preschoolers with Autism Spectrum Disorder. Dig Liver Dis. 2016;48:248–54. 10.1016/J.DLD.2015.11.026. [DOI] [PubMed] [Google Scholar]
  • 170.Houghton R, Ong RC, Bolognani F. Psychiatric comorbidities and use of psychotropic medications in people with autism spectrum disorder in the United States. Autism Res. 2017;10:2037–47. 10.1002/AUR.1848. [DOI] [PubMed] [Google Scholar]
  • 171.Kang DW, Park JG, Ilhan ZE, Wallstrom G, LaBaer J, Adams JB, et al. Reduced incidence of prevotella and other fermenters in intestinal microflora of Autistic children. PLoS ONE. 2013;8:e68322. 10.1371/JOURNAL.PONE.0068322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Ding X, Xu Y, Zhang X, Zhang L, Duan G, Song C, et al. Gut microbiota changes in patients with autism spectrum disorders. J Psychiatr Res. 2020;129:149–59. 10.1016/J.JPSYCHIRES.2020.06.032. [DOI] [PubMed] [Google Scholar]
  • 173.Tomova A, Husarova V, Lakatosova S, Bakos J, Vlkova B, Babinska K, et al. Gastrointestinal microbiota in children with autism in Slovakia. Physiol Behav. 2015;138:179–87. 10.1016/J.PHYSBEH.2014.10.033. [DOI] [PubMed] [Google Scholar]
  • 174.Parracho HMRT, Bingham MO, Gibson GR, McCartney AL. Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children. J Med Microbiol. 2005;54:987–91. 10.1099/JMM.0.46101-0/CITE/REFWORKS. [DOI] [PubMed] [Google Scholar]
  • 175.Zhao RH, Zheng PY, Liu SM, Tang YC, Li EY, Sun ZY, et al. Correlation between gut microbiota and behavior symptoms in children with autism spectrum disorder. Chin J Contemp Pediatr. 2019;21:663–9. 10.7499/J.ISSN.1008-8830.2019.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Strati F, Cavalieri D, Albanese D, De Felice C, Donati C, Hayek J, et al. New evidences on the altered gut microbiota in autism spectrum disorders. Microbiome. 2017. 10.1186/S40168-017-0242-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Rianda D, Agustina R, Setiawan EA, Manikam NRM. Effect of probiotic supplementation on cognitive function in children and adolescents: a systematic review of randomised trials. Benef Microbes. 2019;10:873–82. 10.3920/BM2019.0068. [DOI] [PubMed] [Google Scholar]
  • 178.Liu S, Li E, Sun Z, Fu D, Duan G, Jiang M, et al. Altered gut microbiota and short chain fatty acids in Chinese children with autism spectrum disorder. Sci Rep. 2019. 10.1038/S41598-018-36430-Z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Peng A, Qiu X, Lai W, Li W, Zhang L, Zhu X, et al. Altered composition of the gut microbiome in patients with drug-resistant epilepsy. Epilepsy Res. 2018;147:102–7. 10.1016/J.EPLEPSYRES.2018.09.013. [DOI] [PubMed] [Google Scholar]
  • 180.Olson CA, Vuong HE, Yano JM, Liang QY, Nusbaum DJ, Hsiao EY. The gut microbiota mediates the anti-seizure effects of the ketogenic diet. Cell. 2018;173:1728-1741.e13. 10.1016/J.CELL.2018.04.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.Gómez-Eguílaz M, Ramón-Trapero JL, Pérez-Martínez L, Blanco JR. The beneficial effect of probiotics as a supplementary treatment in drug-resistant epilepsy: a pilot study. Benef Microbes. 2018;9:875–81. 10.3920/BM2018.0018. [DOI] [PubMed] [Google Scholar]
  • 182.Arzani M, Jahromi SR, Ghorbani Z, Vahabizad F, Martelletti P, Ghaemi A, et al. Gut-brain axis and migraine headache: a comprehensive review. J Headache Pain. 2020. 10.1186/S10194-020-1078-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Chen J, Wang Q, Wang A, Lin Z. Structural and functional characterization of the gut microbiota in elderly women with migraine. Front Cell Infect Microbiol. 2020;9:473888. 10.3389/FCIMB.2019.00470/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Partida-Rodríguez O, Serrano-Vázquez A, Nieves-Ramírez ME, Moran P, Rojas L, Portillo T, et al. Human intestinal microbiota: interaction between parasites and the host immune response. Arch Med Res. 2017;48:690–700. 10.1016/J.ARCMED.2017.11.015. [DOI] [PubMed] [Google Scholar]
  • 185.Matsuki T, Tanaka R. Function of the human gut microbiota. In: Marchesi JR, editor. The human microbiota and microbiome. Wallingford: CABI; 2014. p. 90–106. [Google Scholar]
  • 186.Berrilli F, Di Cave D, Cavallero S, D’Amelio S. Interactions between parasites and microbial communities in the human gut. Front Cell Infect Microbiol. 2012;2:141. 10.3389/FCIMB.2012.00141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Verma AK, Verma R, Ahuja V, Paul J. Real-time analysis of gut flora in Entamoeba histolytica infected patients of Northern India. BMC Microbiol. 2012;12:1–11. 10.1186/1471-2180-12-183/FIGURES/6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Partida-Rodríguez O, Brown EM, Woodward SE, Cirstea M, Reynolds LA, Petersen C, et al. Fecal microbiota transplantation from protozoa-exposed donors downregulates immune response in a germ-free mouse model, its role in immune response and physiology of the intestine. PLoS ONE. 2024;19:e0312775. 10.1371/JOURNAL.PONE.0312775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 189.Ramanan D, Bowcutt R, Lee SC, Tang MS, Kurtz ZD, Ding Y, et al. Helminth infection promotes colonization resistance via type 2 immunity. Science. 1979;2016(352):608–12. 10.1126/SCIENCE.AAF3229/SUPPL_FILE/RAMANAN.SM.PDF. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Reynolds LA, Finlay BB, Maizels RM. Cohabitation in the Intestine: Interactions among Helminth Parasites, Bacterial Microbiota, and Host Immunity. J Immunol. 2015;195:4059–66. 10.4049/JIMMUNOL.1501432. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Aboulhoda BE, Abdelfatah M, El-Wakil ES, Alghamdi M, Albadawi EA, Hassan FE, et al. Microbiota-parasite interaction: implication of secretory immunoglobulin A and P2X7 receptor signaling. Discov Med. 2024;36:217–33. 10.24976/DISCOV.MED.202436181.21. [DOI] [PubMed] [Google Scholar]
  • 192.Nieves-Ramírez ME, Partida-Rodríguez O, Laforest-Lapointe I, Reynolds LA, Brown EM, Valdez-Salazar A, et al. Asymptomatic intestinal colonization with protist blastocystis is strongly associated with distinct microbiome ecological patterns. mSystems. 2018. 10.1128/MSYSTEMS.00007-18/SUPPL_FILE/SYS003182239ST7.DOCX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Karpe AV, Beale DJ, Tran CD. Intelligent biological networks: improving anti-microbial resistance resilience through nutritional interventions to understand protozoal gut infections. Microorganisms. 2023;11:1800. 10.3390/MICROORGANISMS11071800. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Gerbaba TK, Gupta P, Rioux K, Hansen D, Buret AG. Giardia duodenalis-induced alterations of commensal bacteria kill Caenorhabditis elegans: a new model to study microbial-microbial interactions in the gut. Am J Physiol Gastrointest Liver Physiol. 2015;308:G550–61. 10.1152/AJPGI.00335.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Koh W, Clode PL, Monis P, Thompson RA. Multiplication of the waterborne pathogen Cryptosporidium parvum in an aquatic biofilm system. Parasit Vectors. 2013;6:1–11. 10.1186/1756-3305-6-270/FIGURES/8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Dwyer DM, Chang KP. Surface membrane carbohydrate alterations of a flagellated protozoan mediated by bacterial endosymbiotes. Proc Natl Acad Sci. 1976;73:852–6. 10.1073/PNAS.73.3.852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Swann J, Jamshidi N, Lewis NE, Winzeler EA. Systems analysis of host–parasite interactions. Wiley Interdiscip Rev Syst Biol Med. 2015;7:381–400. 10.1002/WSBM.1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Wypych TP, Wickramasinghe LC, Marsland BJ. The influence of the microbiome on respiratory health. Nat Immunol. 2019;20:1279–90. 10.1038/s41590-019-0451-9. [DOI] [PubMed] [Google Scholar]
  • 199.Zheng L, Ji Y, Wen X. Fecal microbiota transplantation in the metabolic diseases: current status and perspectives. World J Gastroenterol. 2022;28:2546–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Kumar A, Pramanik J, Goyal N, Chauhan D, Sivamaruthi BS, Prajapati BG, et al. Gut microbiota in anxiety and depression: unveiling the relationships and management options. Pharmaceuticals. 2023. 10.3390/PH16040565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Rahman MM, Islam F, Or-Rashid MH, Mamun AA, Rahaman MS, Islam MM, et al. The gut microbiota (microbiome) in cardiovascular disease and its therapeutic regulation. Front Cell Infect Microbiol. 2022;12:903570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Wang L, Guo G, Xu Y, Li L, Yang B, Zhao D, et al. The effect of fecal microbiota transplantation on antibiotic-associated diarrhea and its impact on gut microbiota. BMC Microbiol. 2024. 10.1186/S12866-024-03261-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Cristofori F, Dargenio VN, Dargenio C, Miniello VL, Barone M, Francavilla R. Anti-inflammatory and immunomodulatory effects of probiotics in gut inflammation: a door to the body. Front Immunol. 2021;12:578386. 10.3389/FIMMU.2021.578386/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Xu T, He X, Chen T. The role of probiotics, postbiotics, and microbial metabolites in preventing and treating chronic diseases. Front Cell Infect Microbiol. 2023;13:1246937. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Alli SR, Gorbovskaya I, Liu JCW, Kolla NJ, Brown L, Müller DJ. The gut microbiome in depression and potential benefit of prebiotics, probiotics and synbiotics: a systematic review of clinical trials and observational studies. Int J Mol Sci. 2022. 10.3390/IJMS23094494/S1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Davani-Davari D, Negahdaripour M, Karimzadeh I, Seifan M, Mohkam M, Masoumi SJ, et al. Prebiotics: definition, types, sources, mechanisms, and clinical applications. 2019. Foods. 10.3390/FOODS8030092. [DOI] [PMC free article] [PubMed]
  • 207.de Costa A. The appendix-mucosal immunity and tolerance in the gut: consequences for the syndromes of appendicitis and its epidemiology. ANZ J Surg. 2022;92:653–60. 10.1111/ans.17522. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Zhao X, Zhao J, Li D, Yang H, Chen C, Qin M, et al. Akkermansia muciniphila: a potential target and pending issues for oncotherapy. Pharmacol Res. 2023;196:106916. 10.1016/J.PHRS.2023.106916. [DOI] [PubMed] [Google Scholar]
  • 209.Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermúdez-Humarán LG, Gratadoux JJ, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A. 2008;105:16731–6. 10.1073/PNAS.0804812105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Mastromarino P, Capobianco D, Miccheli A, Praticò G, Campagna G, Laforgia N, et al. Administration of a multistrain probiotic product (VSL#3) to women in the perinatal period differentially affects breast milk beneficial microbiota in relation to mode of delivery. Pharmacol Res. 2015;95–96:63–70. 10.1016/J.PHRS.2015.03.013. [DOI] [PubMed] [Google Scholar]
  • 211.Flori L, Benedetti G, Martelli A, Calderone V. Microbiota alterations associated with vascular diseases: postbiotics as a next-generation magic bullet for gut-vascular axis. Pharmacol Res. 2024;207: 107334. 10.1016/J.PHRS.2024.107334. [DOI] [PubMed] [Google Scholar]
  • 212.Kim CS. Roles of diet-associated gut microbial metabolites on brain health: cell-to-cell interactions between gut bacteria and the central nervous system. Adv Nutr. 2024;15:100136. 10.1016/J.ADVNUT.2023.10.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Zhu G, Zhao J, Zhang H, Wang G, Chen W. Gut microbiota and its metabolites: bridge of dietary nutrients and Alzheimer’s disease. Adv Nutr. 2023;14:819–39. 10.1016/J.ADVNUT.2023.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Zapolska-Downar D, Siennicka A, Kaczmarczyk M, Kołodziej B, Naruszewicz M. Butyrate inhibits cytokine-induced VCAM-1 and ICAM-1 expression in cultured endothelial cells: the role of NF-κB and PPARα. J Nutr Biochem. 2004;15:220–8. 10.1016/J.JNUTBIO.2003.11.008. [DOI] [PubMed] [Google Scholar]
  • 215.Poll BG, Xu J, Jun S, Sanchez J, Zaidman NA, He X, et al. Acetate, a short-chain fatty acid, acutely lowers heart rate and cardiac contractility along with blood pressure. J Pharmacol Exp Ther. 2021;377:39–50. 10.1124/JPET.120.000187/-/DC1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Haghikia A, Zimmermann F, Schumann P, Jasina A, Roessler J, Schmidt D, et al. Propionate attenuates atherosclerosis by immune-dependent regulation of intestinal cholesterol metabolism. Eur Heart J. 2022;43:518–33. 10.1093/EURHEARTJ/EHAB644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Bell HN, Rebernick RJ, Goyert J, Singhal R, Kuljanin M, Kerk SA, et al. Reuterin in the healthy gut microbiome suppresses colorectal cancer growth through altering redox balance. Cancer Cell. 2022;40:185-200.e6. 10.1016/J.CCELL.2021.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Li Q, Chan H, Liu WX, Liu CA, Zhou Y, Huang D, et al. Carnobacterium maltaromaticum boosts intestinal vitamin D production to suppress colorectal cancer in female mice. Cancer Cell. 2023;41:1450-1465.e8. 10.1016/J.CCELL.2023.06.011. [DOI] [PubMed] [Google Scholar]
  • 219.Acharya A, Shetty SS, Kumari NS. Role of gut microbiota derived short chain fatty acid metabolites in modulating female reproductive health. Human Nutr Metab. 2024;36:200256. 10.1016/J.HNM.2024.200256. [Google Scholar]
  • 220.Abdalkareem Jasim S, Jade Catalan Opulencia M, Alexis Ramírez-Coronel A, Kamal Abdelbasset W, Hasan Abed M, Markov A. The emerging role of microbiota-derived short-chain fatty acids in immunometabolism. Int Immunopharmacol. 2022;110:108983. [DOI] [PubMed] [Google Scholar]
  • 221.Canfora EE, Jocken JW, Blaak EE. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat Rev Endocrinol. 2015;11:577–91. 10.1038/NRENDO.2015.128. [DOI] [PubMed] [Google Scholar]
  • 222.Barandouzi ZA, Lee J, del Carmen RM, Chen J, Henderson WA, Starkweather AR, et al. Associations of neurotransmitters and the gut microbiome with emotional distress in mixed type of irritable bowel syndrome. Sci Rep. 2022;12:1648. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Braga JD, Thongngam M, Kumrungsee T. Gamma-aminobutyric acid as a potential postbiotic mediator in the gut–brain axis. NPJ Sci Food. 2024;8:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Rashid S, Sado AI, Afzal MS, Ahmed A, Almaalouli B, Waheed T, et al. Role of gut microbiota in cardiovascular diseases–a comprehensive review. Ann Med Surg. 2024;86:1483–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Anhê FF, Barra NG, Cavallari JF, Henriksbo BD, Schertzer JD. Metabolic endotoxemia is dictated by the type of lipopolysaccharide. Cell Rep. 2021;36:109691. [DOI] [PubMed] [Google Scholar]
  • 226.Kazemian N, Mahmoudi M, Halperin F, Wu JC, Pakpour S. Gut microbiota and cardiovascular disease: opportunities and challenges. Microbiome. 2020;8:1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Zhen J, Zhou Z, He M, Han H-X, Lv E-H, Wen P-B, et al. The gut microbial metabolite trimethylamine N-oxide and cardiovascular diseases. Front Endocrinol (Lausanne). 2023;14:1085041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Wang L, Wang S, Zhang Q, He C, Fu C, Wei Q. The role of the gut microbiota in health and cardiovascular diseases. Mol Biomed. 2022;3:30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Chaikijurajai T, Wilcox J, Tang WHW. Role of the microbiome in cardiovascular disease. In: Wilkinson MJ, Garshick MS, Taub PR, editors. Prevention and treatment of cardiovascular disease: nutritional and dietary approaches. Cham: Humana; 2021. p. 225–50. [Google Scholar]
  • 230.Zhao Q, Chen Y, Huang W, Zhou H, Zhang W. Drug-microbiota interactions: an emerging priority for precision medicine. Signal Transduct Target Ther. 2023;8:1–27. 10.1038/s41392-023-01619-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Boicean A, Bratu D, Fleaca SR, Vasile G, Shelly L, Birsan S, et al. Exploring the potential of fecal microbiota transplantation as a therapy in tuberculosis and inflammatory bowel disease. Pathogens. 2023;12:1149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Paramsothy S, Kamm MA, Kaakoush NO, Walsh AJ, van den Bogaerde J, Samuel D, et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial. Lancet. 2017;389:1218–28. 10.1016/S0140-6736(17)30182-4. [DOI] [PubMed] [Google Scholar]
  • 233.Borody TJ, Brandt LJ, Paramsothy S. Therapeutic faecal microbiota transplantation: current status and future developments. Curr Opin Gastroenterol. 2014;30:97. 10.1097/MOG.0000000000000027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Cammarota G, Ianiro G, Kelly CR, Mullish BH, Allegretti JR, Kassam Z, et al. International consensus conference on stool banking for faecal microbiota transplantation in clinical practice. Gut. 2019;68:2111–21. 10.1136/GUTJNL-2019-319548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Cryan JF, Dinan TG. Mind-altering microorganisms: the impact of the gut microbiota on brain and behaviour. Nat Rev Neurosci. 2012;13:701–12. 10.1038/nrn3346. [DOI] [PubMed] [Google Scholar]
  • 236.Arnold J, Glazier J, Mimee M. Genetic engineering of resident bacteria in the gut microbiome. J Bacteriol. 2023;205:e00127-e223. 10.1128/JB.00127-23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 237.Yao L, Seaton SC, Ndousse-Fetter S, Adhikari AA, DiBenedetto N, Mina AI, et al. A selective gut bacterial bile salt hydrolase alters host metabolism. Elife. 2018;7:e37182. 10.7554/ELIFE.37182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 238.Guo CJ, Allen BM, Hiam KJ, Dodd D, van Treuren W, Higginbottom S, et al. Depletion of microbiome-derived molecules in the host using Clostridium genetics. Science. 2019;366:eaav1282. 10.1126/SCIENCE.AAV1282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 239.Funabashi M, Grove TL, Wang M, Varma Y, McFadden ME, Brown LC, et al. A metabolic pathway for bile acid dehydroxylation by the gut microbiome. Nature. 2020;582:566. 10.1038/S41586-020-2396-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 240.Deriu E, Liu JZ, Pezeshki M, Edwards RA, Ochoa RJ, Contreras H, et al. Probiotic bacteria reduce salmonella typhimurium intestinal colonization by competing for iron. Cell Host Microbe. 2013;14:26–37. 10.1016/J.CHOM.2013.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Schultz M, Clinical use of E. coli Nissle,. in inflammatory bowel disease. Inflamm Bowel Dis. 1917;2008(14):1012–8. 10.1002/IBD.20377. [DOI] [PubMed] [Google Scholar]
  • 242.Sonnenborn U. Escherichia coli strain Nissle 1917-from bench to bedside and back: history of a special Escherichia coli strain with probiotic properties. FEMS Microbiol Lett. 2016. 10.1093/FEMSLE/FNW212. [DOI] [PubMed] [Google Scholar]
  • 243.Charbonneau MR, Isabella VM, Li N, Kurtz CB. Developing a new class of engineered live bacterial therapeutics to treat human diseases. Nat Commun. 2020;11:1–11. 10.1038/s41467-020-15508-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 244.Nakamura T, Sasaki T, Fujimori M, Yazawa K, Kano Y, Amano J, et al. Cloned cytosine deaminase gene expression of Bifidobacterium longum and application to enzyme/pro-drug therapy of hypoxic solid tumors. Biosci Biotechnol Biochem. 2002;66:2362–6. 10.1271/BBB.66.2362. [DOI] [PubMed] [Google Scholar]
  • 245.Yi C, Huang Y, Guo ZY, Wang SR. Antitumor effect of cytosine deaminase/5-fluorocytosine suicide gene therapy system mediated by Bifidobacterium infantis on melanoma. Acta Pharmacol Sin. 2005;26:629–34. 10.1111/J.1745-7254.2005.00094.X. [DOI] [PubMed] [Google Scholar]
  • 246.Li R, Helbig L, Fu J, Bian X, Herrmann J, Baumann M, et al. Expressing cytotoxic compounds in Escherichia coli Nissle 1917 for tumor-targeting therapy. Res Microbiol. 2019;170:74–9. 10.1016/J.RESMIC.2018.11.001. [DOI] [PubMed] [Google Scholar]
  • 247.Pahle J, Menzel L, Niesler N, Kobelt D, Aumann J, Rivera M, et al. Rapid eradication of colon carcinoma by Clostridium perfringens Enterotoxin suicidal gene therapy. BMC Cancer. 2017. 10.1186/S12885-017-3123-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Wang X, Peng J, Cai P, Xia Y, Yi C, Shang A, et al. The emerging role of the gut microbiota and its application in inflammatory bowel disease. Biomed Pharmacother. 2024;179:117302. 10.1016/J.BIOPHA.2024.117302. [DOI] [PubMed] [Google Scholar]
  • 249.Ando H, Lemire S, Pires DP, Lu TK. Engineering modular viral scaffolds for targeted bacterial population editing. Cell Syst. 2015;1:187–96. 10.1016/J.CELS.2015.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 250.Federici S, Kredo-Russo S, Valdés-Mas R, Kviatcovsky D, Weinstock E, Matiuhin Y, et al. Targeted suppression of human IBD-associated gut microbiota commensals by phage consortia for treatment of intestinal inflammation. Cell. 2022;185:2879-2898.e24. 10.1016/J.CELL.2022.07.003. [DOI] [PubMed] [Google Scholar]
  • 251.Rahman M, Wadud M, Islam T, Hussain M, Bristy E, Tuhin A. Evaluation of antibacterial activity of piper betel leaves and nigella sativa seeds against multidrug resistant food and water borne pathogenic bacteria: an in vitro study model. Microbiol Res J Int. 2018;22:1–11. 10.9734/MRJI/2017/37796. [Google Scholar]
  • 252.Sultana A, Saha O, Rahman Sid A, Saha A, Hussain MS, Islam T. Molecular detection of multidrug resistance pathogenic bacteria from protective materials used by Healthcare Workers (HCW); Bangladesh scenario. J Appl Sci. 2018;18:48–55. 10.3923/JAS.2018.48.55. [Google Scholar]
  • 253.Ismael NM, Azzam M, Abdelmoteleb M, El-Shibiny A. Phage vB_Ec_ZCEC14 to treat antibiotic-resistant Escherichia coli isolated from urinary tract infections. Virol J. 2024;21:1–16. 10.1186/S12985-024-02306-0/FIGURES/9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Febvre HP, Rao S, Gindin M, Goodwin NDM, Finer E, Vivanco JS, et al. PHAGE study: effects of supplemental bacteriophage intake on inflammation and gut microbiota in healthy adults. Nutrients. 2019;11:666. 10.3390/NU11030666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Gindin M, Febvre HP, Rao S, Wallace TC, Weir TL. Bacteriophage for Gastrointestinal Health (PHAGE) study: evaluating the safety and tolerability of supplemental bacteriophage consumption. J Am Coll Nutr. 2019;38:68–75. 10.1080/07315724.2018.1483783. [DOI] [PubMed] [Google Scholar]
  • 256.Kumar P, Kiran S, Saha S, Su Z, Paulsen T, Chatrath A, et al. ATAC-seq identifies thousands of extrachromosomal circular DNA in cancer and cell lines. Sci Adv. 2020. 10.1126/SCIADV.ABA1590/SUPPL_FILE/ABA1590_SM.PDF. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Ichikawa M, Nakamoto N, Kredo-Russo S, Weinstock E, Weiner IN, Khabra E, et al. Bacteriophage therapy against pathological Klebsiella pneumoniae ameliorates the course of primary sclerosing cholangitis. Nat Commun. 2023;14:1–13. 10.1038/s41467-023-39029-9. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Citations

  1. Davani-Davari D, Negahdaripour M, Karimzadeh I, Seifan M, Mohkam M, Masoumi SJ, et al. Prebiotics: definition, types, sources, mechanisms, and clinical applications. 2019. Foods. 10.3390/FOODS8030092. [DOI] [PMC free article] [PubMed]

Data Availability Statement

No datasets were generated or analysed during the current study.


Articles from Gut Pathogens are provided here courtesy of BMC

RESOURCES