Skip to main content
Springer Nature - PMC COVID-19 Collection logoLink to Springer Nature - PMC COVID-19 Collection
. 2022 Oct 29;31(12):3603–3615. doi: 10.1007/s00586-022-07429-y

An unexpected connection: A narrative review of the associations between Gut Microbiome and Musculoskeletal Pain

Valerio Tonelli Enrico 1,2,, Nam Vo 1, Barbara Methe 3, Alison Morris 3, Gwendolyn Sowa 1,4
PMCID: PMC9617047  PMID: 36308543

Abstract

Purpose

Multiple diverse factors contribute to musculoskeletal pain, a major cause of physical dysfunction and health-related costs worldwide. Rapidly growing evidence demonstrates that the gut microbiome has overarching influences on human health and the body’s homeostasis and resilience to internal and external perturbations. This broad role of the gut microbiome is potentially relevant and connected to musculoskeletal pain, though the literature on the topic is limited. Thus, the literature on the topic of musculoskeletal pain and gut microbiome was explored.

Methods

This narrative review explores the vast array of reported metabolites associated with inflammation and immune-metabolic response, which are known contributors to musculoskeletal pain. Moreover, it covers known modifiable (e.g., diet, lifestyle choices, exposure to prescription drugs, pollutants, and chemicals) and non-modifiable factors (e.g., gut architecture, genetics, age, birth history, and early feeding patterns) that are known to contribute to changes to the gut microbiome. Particular attention is devoted to modifiable factors, as the ultimate goal of researching this topic is to implement gut microbiome health interventions into clinical practice.

Results

Overall, numerous associations exist in the literature that could converge on the gut microbiome’s pivotal role in musculoskeletal health. Particularly, a variety of metabolites that are either directly produced or indirectly modulated by the gut microbiome have been highlighted.

Conclusion

The review highlights noticeable connections between the gut and musculoskeletal health, thus warranting future research to focus on the gut microbiome’s role in musculoskeletal conditions.

Keywords: Gut microbiome, Musculoskeletal pain, Inflammation, Homeostasis, Pain modulation

Introduction

The complex and multi-dimensional experience of pain involves various mechanisms, which are only partially understood. The relationship between gut microbiome (GM) and painful conditions has received increasing research attention. Growing evidence shows that GM is a crucial modulator of human physiological homeostasis, playing a not-fully-understood but undoubtedly significant role in systemic inflammation, immunity, circadian rhythm, and regulation of hormone levels; all these aspects of homeostasis have been linked to pain. GM has been associated with visceral pain [1], inflammatory pain, headache, neuropathic pain, chronic pain, and opioid tolerance [2]. With contributions from multiple types of pain, musculoskeletal (MSK) pain is highly prevalent [3], a significant source of disability [4], and a frequent reason for medical care [5]. There is initial evidence linking GM and MSK pain, though the mechanisms are not clarified yet. This review covers the current literature on the common mechanisms of GM and pain modulation, focusing on potential connections between GM and MSK pain mechanisms to help direct further research on the topic.

The composition of the GM is complex and spans three kingdoms (bacteria, archaea, fungi), and it has recently been expanded to include viruses [6]. In 2012, a paper pointed at the number of known species composing the GM to be over 1000 [7], while a more recent effort (2019) to study the composition raised that number to almost 2000 species [8].

The overall genomic material from intestinal microbes has been estimated to be greater than 100 times the size of the human genome [9]. While more than 99% of the GM bacteria are from only four phyla (Firmicutes, Bacteroidetes, Proteobacteria, and Actinobacteria), most bacteria in healthy adults come from Firmicutes or Bacteroidetes [10]. It is currently postulated that the microbiome population is shaped through adaptation that likely involves factors on multiple levels [11] (personal, interpersonal, environmental, and geographical).

GM samples for analysis are collected from stool, and microbiome profiling is conducted using, in most cases, culture-independent methods to sequence16S rRNA gene amplicons or shotgun sequencing of whole microbial communities (metagenomics). Other methods for studying the GM population exist, such as whole microbial community RNA sequencing (metatranscriptomics), while proteomics and metabolomics can be used to obtain simultaneous information about the host and microbiome [12]. The choice of method to use is often dictated by funding and technology, as higher costs and more complex methodologies accompany more thorough analyses.

Gut microbiome and pain

Gut microbiome homeostasis and disruption

In healthy individuals, GM is characterized by a state of dynamic homeostasis defined by the richness and diversity of the GM population [13] and by its stability and resilience through perturbations of various types [14]. Numerous modifiable and non-modifiable factors play a role in determining the GM composition and its alterations (Table 1). In physiological homeostasis, the GM has been hypothesized to play a role in properly maintaining the permeability of the intestinal wall [15]. Such permeability is fundamental in how numerous antigens (from foods, pathogens, and gut microbes) are treated in the gut and how they affect the immune response [16].

Table 1.

Known contributing factors to GM composition and alteration

Examples References
Non-modifiable factors
Gut architecture Microphysical gut structure [17]
Macrophysical gut structure [17]
Human genetics FUT2 [18]
NOD2 [19]
LCT [20]
Birthing history Vaginal delivery [21]
C-section [22]
Early feeding patterns Breastfeeding [23]
Formula [24]
Age (or life stages) Children, [25]
Adults, [26]
Elderly [27]
Geographic location Climate, [28]
Lifestyles, [29]
Genetic background, [30, 31]
Dwelling location, [32]
Food access, [33]
Employment [34]
Members of minorities Ethnicity [35, 36]
Self-identified race [33]
Sexual identity [33]
Gender status [33]
Modifiable factors
Diet Dietary patters
Western [37]
Mediterranean [38]
Pro- or anti-inflammatory dietary tendencies [39]
Lifestyle choices Smoking, [40]
Alcohol consumption, [41]
Exercise [42]
Traumatic events Traumatic brain injury, [43, 44]
Spinal cord injury, [45, 46]
Chronic stress [47, 48]
Interventions Probiotic, [49]
Prebiotic, [50]
Fecal microbiota transplant (FMT) [51]
Prescription Drugs Antibiotics, [52]
Proton pump inhibitors, [53]
Laxatives, [54]
Metformin [55]
Exposure to chemicals pollutants, or drugs Cadmium chloride, [56]
Arsenite, [57]
glyphosate [58]

Perturbations to a healthy GM ecosystem can result in dysbiosis, a distinct ecological state of the microbial community [59]. Dysbiosis could involve changes to critical bacterial species, leading to altered permeability, physiological and metabolic functions, and ultimately predisposing diseases. An example of dysbiosis is the so-called “leaky gut syndrome”, a condition characterized by altered permeability of the intestinal walls to antigens [60], leading to systemic inflammation and aberrant immune response [61]. Other examples of diseases characterized by altered and specific gut microbiome blueprints include neurodegenerative diseases [62], metabolic syndrome [63], and inflammatory bowel disease [10].

In the case of painful conditions, a handful of specific species correlates with disease status. One example includes Faecalibacterium Prausnitzii (FP), whose lower concentrations correlate with osteoarthritis severity in older female adults [64], and Crohn’s disease (a condition for which pain is the most common symptom) [65]. In line with these findings, higher concentrations of FP correlate with anti-nociceptive effects in a rat model of Irritable Bowel Disease [66]. Other species relevant to painful conditions are the Streptococcus species, with an increase associated with knee osteoarthritis pain [67], or the Coprococcus species, the depletion of which has been associated with chronic widespread pain [68].

While individual species have been implicated in many conditions, overall evidence highlights that interactions among different species are complex and more likely to explain the pathophysiology of various diseases (including painful ones) than any single species alone. For example, a recently published study found that a combination of species correctly predicted the diagnosis of 88% of fibromyalgia patients [69].

Gut microbiome, inflammation, immune response, and impact on host physiology

The current evidence converges on the crucial role of GM in mediating systemic inflammation. In a state of inflammation (both local and systemic), the body can change how stimuli are perceived and processed. For example, in an inflammatory state, noxious stimuli can produce an increased response to pain, known as hyperalgesia, while non-noxious stimuli can be perceived as painful, known as allodynia [70]. The state of low-grade chronic systemic inflammation has been theorized to predispose to chronic pain and other chronic diseases [71].

Converging evidence highlights that GM can impact the inflammatory state by producing various metabolites (explained in the next section) and modulating systemic inflammatory cytokines production. For example, a study on healthy subjects linked the GM composition to the capacity to produce inflammatory cytokines (IL-1b, IL-6, TNFα, IFNγ, IL-17, and IL-22) [72]. A recent study on cytokine levels in SARS-CoV-2 patients highlighted that cytokine levels mediated by the GM composition were associated with the magnitude of the COVID-19 severity [73]. Another study on mice suggested GM modulating specific cytokines by affecting T cells [74].

Several reviews also emphasized the proven link between GM and immune-mediated inflammatory response. The modulatory changes that the GM has on the immune response are mediated by changes to the function of B and T cells, which also influences the host’s resistance to pathogens [75]. These immune function changes also impact the various GM-mediated cytokines and chemokines levels’ (e.g., IL-1, IL-6, IL-10, TGFβ, TNFα, IFNγ, IL-17, and IL-22). Such changes have been related to a variety of alterations of physiological homeostasis, including metabolic function (insulin resistance and obesity [76], exercise-induced stress behavior and its impact on the gut-microbiota-brain axis [77], and cancer [78]), modulation of nerve function (blood–brain barrier integrity and brain health [79]), and autoimmunity [80].

Gut microbiome metabolites as possible contributors to pain

The GM produces a broad spectrum of metabolites that impact the human body on multiple levels (Table 2), from a local impact on the GI tract to organ-specific distal from the GI tract to systemic [81]. The GM’s multiple metabolites that correlate with pain will be addressed in this section to highlight the possible involvement of specific pathways and mechanisms that should be investigated in future studies and could become relevant for potential therapies.

Table 2.

GM metabolites as possible contributing mechanisms of pain

Hormones (or compounds acting as hormones) References
Direct effect from GM Short chain fatty acids  Butyrate [69, 82]
 Propionate [69, 82]
Neurotransmitters  Serotonin [83]
 Dopamine [84, 85]
 Noradrenaline [86]
 Glutamate [87, 88]
 GABA [87, 88]
Indirect effect from GM Hypothalamic–pituitary–adrenal axis (HPA)  Cortisol [89]
GI hormones  Leptin [9092]
 Ghrelin [93]
Vitamins and nutrients References
D [94, 95]
B1-Thiamine [96]
B2-Riboflavin
B3-Nicotinic acid
B5-Pantothenic acid
B6-Pyridoxine
B7-Biotin
B9-Folic acid
B12-Cyanocobalamin [96, 97]
para-aminobenzoic acid [96]
Inositol
choline

GM in humans produces various hormones (or compounds acting as such), so GM can be considered an endocrine organ functionally [98]. This definition relies on the fact that GM produces numerous chemicals that act via the bloodstream to distal sites in the human body. These hormones can be classified as being directly produced by the GM or indirectly regulated by the GM.

Hormones (and compounds acting as hormones) directly produced by the gut microbiome

The first group includes chemicals that mediate pain, including short-term fatty acids (SCFAs) and neurotransmitters (including some precursors). SCFAs are produced in the large intestine by the GM through anaerobic fermentation of dietary fibers [99]; bacteria from the Bacteroidetes phylum mainly produce propionate, while bacteria from the Firmicutes phylum mainly produce butyrate [82]. SCFAs play a critical role in modulating intestinal inflammation and epithelial barrier function [82]; dysfunction at the epithelial barrier level is one of the leading causes of “leaky gut syndrome” that plays a vital role in various painful conditions. Moreover, SCFAs produced in the gut can enter the bloodstream [82], further contributing to the modulation of systemic inflammation [81]. For example, butyrate and propionate serum levels were significantly altered in a group of subjects affected by fibromyalgia compared to healthy controls [69].

Serotonin, dopamine, noradrenaline, glutamate, and GABA are directly produced by the GM or indirectly regulated by it. Dopamine, long considered a key modulator of reward-seeking behavior, is produced in large quantities in the gut [100]; it also participates in the immune response [101], thus being important in the modulation of systemic inflammation. Interestingly, at least 50% of dopamine is synthesized in the gut, with GM likely playing a significant part in the production process [102]. Its complex role in the human body includes a modulatory function for chronic pain through the nigrostriatal and mesolimbic pathways [103]. Also, growing evidence has been mounting on the implications of dopamine dysregulation in the brain as a pivotal player in chronic pain’s sensory and affective aspects [84]. A review of mice studies strongly suggested that alterations in the GM significantly compromise dopaminergic neurotransmission in the brain [85].

Serotonin, a complex neuromodulator that regulates various processes [104], including mood regulation [105], learning and memory skills [106], and various other physiological processes, is estimated to be produced 90% in the gut [107]. Of specific importance for pain, serotonin plays a crucial role in the gut-brain axis [108], connecting the central nervous system and the gastrointestinal tract bidirectionally [109]. The decades-old concept of the gut-brain axis has recently been broadened to include the GM in the so-called gut-brain-microbiome axis [110] (or the microbiota-gut axis [111]); this more inclusive concept owes to the growing evidence of the GM’s role in modulating the gut-brain pathway [112]. For example, GM (through various species, including Clostridia, Bacteroides, and Escherichia [113]) can alter serotonin levels by affecting the production of tryptophan, serotonin’s precursor [114]. Moreover, the brain-gut microbiome axis modulates other different physiological functions associated with pain, such as modulation of the systemic inflammation in various mice models [115], alteration of the GI tract function [116] and directly impacting different kinds of painful syndromes [83] in humans.

The production of noradrenaline has been associated with both specific species (Escherichia, Saccharomyces, and Bacillus [117]) and alpha diversity levels in different studies [118]. This key neurotransmitter plays a role in pain modulation [86] and analgesia [119]. Moreover, noradrenaline plays a definite role in the mediation of inflammation [120], participating in both the acute and the chronic regulation of the immune response [121] through its effect on the autonomic nervous system [122] and regulation of the circadian rhythm [123].

Glutamate and GABA, functioning respectively as the primary excitatory [124] and inhibitory [125] neurometabolites in the central nervous system, play various key physiological roles (e.g., inflammation [126]), including the processing and modulation of pain [87]. Alterations in the metabolism of both molecules have been associated with chronic pain [88]. Various gut microbes participate in the production of both these metabolites. For example, certain species have been found to directly produce glutamate (Lactobacillus and Bifidobacterium [127]), whereas Bacteriodes have been associated with GABA production and related specific brain signatures typical of depression [128].

Hormones indirectly produced by the gut microbiome

The second group of indirect regulation of hormones by the GM includes cortisol (chief stress hormone in the hypothalamic–pituitary–adrenal axis (HPA) [129]) and the GI hormones ghrelin and leptin, which are also relevant mediators and modulators of pain. Cortisol is the principal stress hormone in humans [130]. While it does not directly affect pain [131], a chronic cortisol dysfunction can negatively impact the body’s ability to cope with prolonged stress. Maladaptive responses to stress can perpetuate widespread inflammation and pain, potentially sustaining mechanisms predisposing to chronic pain [89]. In mice models, specific pathogens in the GM (e.g., E. Coli infection) can heighten the HPA activity levels, thus driving the stress response higher than in cases where the GM is free of such pathogens [132].

The GI hormone ghrelin has an anti-inflammatory effect by increasing the level of anti-inflammatory cytokines in the serum. Rat-model studies exploring the role of ghrelin on pain showed its anti-nociceptive effects on acute pain [133], neuropathic pain [134], and inflammatory pain [135]. In humans, evidence points to a modulatory effect of GM on the serum levels of ghrelin [93]. Though this evidence is not conclusive, it also points to a participating role of ghrelin on the gut-brain axis when dealing with stress [136], furthering its potential role in mediating pain.

Leptin participates in the pathogenesis of neuropathic pain [90], but it also has a role in immunometabolic inflammation, characteristic of many chronic pain conditions [91]. Moreover, a study suggests that serum concentration of leptin may be a promising biomarker for predicting acute pain transition to chronic [92]. The effect of leptin on inflammation and immunometabolism makes it a potential candidate for the therapeutic development of novel interventions for treating autoimmunity [137] and metabolic diseases [138]. In rat models, the number of specific gut microbial species populations correlates with the modulation of serum levels of leptin [139]. In humans, the GM indirect modulation of leptin appears to be regulated through the production of other metabolites, such as SCFAs [140].

Vitamins and nutrients produced by the gut microbiome

A healthy GM contributes to the production of various vitamins and nutrients [141], the lack of many of which have been associated with pain (e.g., Vitamins of the B group and Vitamin D).

There is a well-established body of evidence that many microbes that reside in the gut produce vitamins of the B group [142]. Among the group B vitamins, the largest body of literature focuses on the positive effects of vitamin B12 in painful conditions, particularly for decreased low back pain and neuralgia [97]. The evidence for the other vitamins that are part of the B group is weak and unable to differentiate between individual vitamins' effects because these vitamins are most often administered together in available studies. For example, a Cochrane review from 2008 [96] highlighted that the vitamin B group positively affects neuropathic pain and patients' functional level. Overall, the analgesic effect of the group B vitamins could be explained by anti-inflammatory, anti-nociceptive, and neuroprotective effects widely described in the literature.

For vitamin D, the evidence is less strong. While there is evidence that vitamin D levels in the plasma affect the gut microbiome [143], no evidence could be found of the reverse association. Significant literature points to the association between vitamin D and pain severity. A systematic review and meta-analysis of observational studies pointed at a significant association between vitamin D and low back pain [144], though it did not find an association between this vitamin and pain intensity. Moreover, two other reviews found that vitamin D supplementation has a meaningful, beneficial effect of reducing pain in patients experiencing chronic pain [94] and chronic widespread pain [95] while being safe. While the mechanisms behind this pain-decreasing effect are currently unclear, it has been postulated that they may have to do with the anti-inflammatory effect of vitamin D [94].

Other metabolites produced by the gut microbiome with a potential indirect effect on pain

A growing body of evidence suggests that epigenetics and mRNA expression are altered in pain subjects [97]. Various metabolites produced by the GM have an active role in epigenetics, for example, acetyl-CoA [145], lactate [146], glycine [147], serine [148], and methionine [149]. Moreover, preliminary evidence points to a potential role for Acetyl-CoA and lactate in the immune and inflammatory responses [150, 151]. Interestingly, specific B vitamins (including B2, B3, B5, B6, B9, and B12) also participate in regulating genetic responses to environmental factors (epigenetics) through the modulation of chromatin-modulating enzymes [152]. Overall, the presence of these currently poorly understood mechanisms points to the overarching complexity and involvement of multiple systems in the painful experience.

Gut microbiome and specific pain syndromes

Inflammatory and neuropathic pain

A study on inflammatory pain [153] found that nociceptive pain signal is decreased in germ-free mice compared to conventional mice. The authors concluded that the interactions between microbiome and host are essential in modulating inflammatory pain. Another study [154] highlighted that pernicious changes in the GM of mice correlate with temporomandibular joint (TMJ) inflammatory pain. Subsequent improvement of GM through fecal transplantation caused a reduction in such pain, strongly suggesting that careful manipulation of the GM has the potential to be used as a therapeutic approach.

Neuropathic pain, caused by a lesion to a nerve of the somatosensory nervous system, has also been impacted by GM-mediated alterations of inflammatory metabolites in rats [155]. Another study [156] used a chronic constriction injury model of neuropathic pain on mice and found that GM modulates neuropathic pain by affecting T cell-mediated immune and inflammatory responses. This finding underlines the relevance of the GM to the modulation of pain even when pain is mechanically induced. Another study on adults affected by HIV experiencing neuropathic pain found that gut dysbiosis-driven loss of alpha diversity in GM was significantly associated with peripheral neuropathic pain [157]. The authors described that increased pro-inflammatory and decreased anti-inflammatory GM metabolites could explain these results.

Pain perception and generalized pain

Chronic pain and Fibromyalgia

Chronic widespread pain (CWP) has been characterized by decreased alpha diversity of the GM. In a study on a chronic widespread pain population, Coprococcus comes was the most significantly reduced species in the GM [68]. A study also found that the alpha diversity of GM is reduced in fibromyalgia patients [158]. This study's participants significantly reduced the population of bacteria from the Bifidobacterium and Eubacterium genera. This finding is interesting because both of these genera participate in the metabolism of neurotransmitters in the host. Further analysis of the serum metabolome discovered significant changes in the levels of glutamate and serine. The authors concluded that the changes in the GM were associated with changes in neurotransmitter metabolism, which contributed to fibromyalgia's clinical presentation.

Pain perception

Based on the framework that correlates the gut-brain axis to various diseases, a recent study showed that acute pain perception and anxiety state were associated with specific species within the GM of young, healthy males [159]. Specifically, pain sensitivity showed a positive association with bacteria from the Firmicutes phylum and a negative association with bacteria from the Bacteroidetes phylum. Moreover, a correlation was observed between anxiety state and the Bifidobacterium genus. Nonetheless, acute pain perception and anxiety were not directly correlated.

Another study that looked at stool classification to ascertain GM's status found that stool consistency was a significant predictor of pain perception in healthy subjects [160]. This study used the Bristol Stool Form Scale (a widely used diagnostic medical tool designed to classify human stool into seven categories) and found a significant and positive association with pain and anxiety. Even though this study did not look directly at the correlation between GM and Bristol Stool Form Scale, other studies did [161], finding the associations significant and worthy of inclusion in future studies on GM.

Opioid tolerance

The opioid epidemic affecting the US has severe repercussions on individuals and society. On the one hand, the long-term use of opioids for various pain conditions has been associated with adverse changes in the GM; on the other hand, the GM may predispose and even mediate the opioid tolerance that results from chronic exposure to opioids [162]. From an individual's health perspective, opioid use and gut microbiome health interaction seem reciprocally interactive.

The mechanisms involved have been postulated to vary from opioid use, possibly inducing gut dysbiosis, disrupting the gut barrier (resulting in the "leaky gut syndrome" mentioned above), and facilitating the translocation of pernicious bacteria. Other hypothesized mechanisms include initiating Toll-Like Receptors (TLRs)-mediating gut inflammation (with concurrent releasing of pro-inflammatory cytokine), and regulating neuronal excitability in the peripheral nervous system [2].

GM and musculoskeletal pain

OA pain

Though various reviews could be found on GM and OA [163, 164], only one considered the impact of GM composition on pain symptoms [165]. Sánchez Romero et al., who reviewed GM and OA pain, found only three studies on the subject. Overall, the review highlighted weak evidence for the link between GM and OA pain, mainly due to the lack of high-quality research.

A Dutch study highlighted significant correlations between GM composition and knee OA [166]. The results were significant after adjusting for smoking, alcohol intake, and BMI. A more prominent presence of Streptococcus species in the subjects' stool samples was associated with more severe knee osteoarthritis (assessed through joint effusion) and increased OA knee pain. The results were replicated using a different Dutch cohort; Streptococcus species were also significantly associated with OA pain in this case. The authors were able to determine that the inflammation localized at the knee joint was driving the link between GM and pain; thus, they concluded that GM could be a potential way to treat OA-driven knee pain,

Another study compared symptomatic vs. asymptomatic hand OA in a large cohort (n = 1388) [167]. It found differences in the relative abundance of specific species, such as a low relative abundance of the genus Roseburia and a high relative abundance of the genera Biophila and Desulfovibrio in the symptomatic hand pain group. The study also found alterations in functional pathways relative to amino acids, carbohydrates, and lipid metabolism. Overall, the authors concluded that GM-related metabolic dysfunction might play a part in how systemic inflammation links to OA's pain symptoms.

A prior study examined GM after a 12-week supplementation of Green-lipped mussel extract or glucosamine sulfate [168] The consumption of either compound did not significantly correlate with changes in the GM; however, it showed decreases in various species associated with higher inflammatory status (e.g., Clostridia and Staphylococcus) and an increase in species associated with decreased inflammatory status (e.g., Lactobacillus and Eubacterium). The authors concluded that GM should be considered when studying the pathogenesis and treatment of musculoskeletal conditions.

LBP obese subjects

Only one study explored the association between back pain and GM composition [169]. The study looked at a cohort of 36 obese subjects with (n = 14) or without (n = 22) LBP, a subset of a larger trial on Vitamin D supplementation. The study found significant differences in specific bacterial genera between groups; Adlercreutzia, Roseburia, and Uncl. Christensenellaceae were significantly more abundant in LBP participants, while Dialister and Lactobacillus were more abundant among those without LBP. The authors also concluded that increased inflammation could have been the cause for such association, thus suggesting an underlying mechanism found elsewhere in scientific literature.

Conclusion

No single gut species or combination of gut species has been consistently associated with health conditions across different host populations. This inconsistency may be due to multiple causes, the foremost of which is that the literature on the topic is relatively recent and lacks comprehensive and systematic studies to understand this complex area of research fully. For example, it could be that the changes in microbiome species across different populations, ages, genetic backgrounds, and lifestyles converge into different patterns of pathological microbiota populations. These complex interactions would require large longitudinal studies to be addressed and are challenging to run for multiple reasons, including costs, time demands, and participant attrition.

Furthermore, it could be that some subsets of affected host populations may be more prone than others to changes in microbiota composition leading to pathological outcomes; this possibility could be mediated by a variety of mechanisms, including genetic predispositions and overall health status. If this is the case, the complexity of researching the topic will require access to large datasets and, potentially, broad cooperation among research groups to power studies large enough to address the complexity of the subject related to health and disease. Last, it could be that the intricacy of the interaction is due to a bidirectional, self-feeding pattern. In this case, an initial perturbation of the GM could lead to obesity and changes in diet, leading to pain and intolerance to physical activity, thus further exacerbating GM alterations.

In conclusion, research is still nascent on the potential role of GM in MSK pain, as evidenced by the scarcity of literature on this topic; nonetheless, a good amount of scientific research highlights numerous possible mechanistic contributors to this association, thus justifying further investigation. This combination of scarce literature and multiple plausible mechanisms raises several questions and strongly encourages more research in the area. Such research should answer questions on the association between GM and musculoskeletal pain and if this relationship is causal. Moreover, it should assess the specific GM changes that drive MSK pain and what mechanisms and therapeutic targets could be identified for clinical interventions. Ideally, a combination of larger systematic cohort studies and clinical trials could help determine these associations and their feasibility for implementation in clinical practice.

Acknowledgements

The authors would like to thank Jessa Darwin for editorial assistance on the manuscript.

Authors contributions

VTE conceptualized and designed the study, performed the literature review and collected data from it, analyzed and synthetized the content from the literature review, critically revised the manuscript for important intellectual content. GS conceptualized and designed the study, interpreted and synthetized the content from the literature review, critically revised the manuscript for important intellectual content. NV interpreted and synthetized the content from the literature review, critically revised the manuscript for important intellectual content. BM interpreted and synthetized the content from the literature review, critically revised the manuscript for important intellectual content. AM interpreted and synthetized the content from the literature review, critically revised the manuscript for important intellectual content. All authors gave final approval for publication and agreed to be accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Funding

The first and corresponding author, Valerio Tonelli Enrico, had the time used in performing this research covered by a supplemental grant (U19AR076725-01S1) that is part of the Back Pain Consortium (BACPAC) consortium. The BACPAC Research Program is administered by the National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS). This research was supported by the National Institutes of Health through the NIH HEAL Initiative under award number 1U19AR076725-01. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or its NIH HEAL Initiative.

Declarations

Conflict of interest

All other authors have no competing interests to declare that are relevant to the content of this article.

Footnotes

Publisher's Note

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

References

  • 1.Moloney RD, Johnson AC, O’Mahony SM, et al. Stress and the microbiota-gut-brain axis in visceral pain: relevance to irritable bowel syndrome. CNS Neurosci Ther. 2016;22:102–117. doi: 10.1111/cns.12490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Guo R, Chen LH, Xing C, Liu T. Pain regulation by gut microbiota: molecular mechanisms and therapeutic potential. Br J Anaesth. 2019;123:637–654. doi: 10.1016/j.bja.2019.07.026. [DOI] [PubMed] [Google Scholar]
  • 3.Luque-Suarez A, Martinez-Calderon J, Falla D. Role of kinesiophobia on pain, disability and quality of life in people suffering from chronic musculoskeletal pain: a systematic review. Br J Sports Med. 2019;53:554–559. doi: 10.1136/bjsports-2017-098673. [DOI] [PubMed] [Google Scholar]
  • 4.Vos T, Flaxman AD, Naghavi M, et al. Years lived with disability (YLDs) for 1160 sequelae of 289 diseases and injuries 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380:2163–2196. doi: 10.1016/S0140-6736(12)61729-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hawker GA. The assessment of musculoskeletal pain. Clin Exp Rheumatol. 2017;35:S8–S12. [PubMed] [Google Scholar]
  • 6.Peroni DG, Nuzzi G, Trambusti I, et al. Microbiome composition and its impact on the development of allergic diseases. Front Immunol. 2020;11:700. doi: 10.3389/FIMMU.2020.00700/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.D’Argenio V, Salvatore F. The role of the gut microbiome in the healthy adult status. Clin Chim Acta. 2015;451:97–102. doi: 10.1016/j.cca.2015.01.003. [DOI] [PubMed] [Google Scholar]
  • 8.Almeida A, Mitchell AL, Boland M, et al. A new genomic blueprint of the human gut microbiota. Nature. 2019;568:499–504. doi: 10.1038/s41586-019-0965-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Putignani L, Del Chierico F, Petrucca A, et al. The human gut microbiota: a dynamic interplay with the host from birth to senescence settled during childhood. Pediatr Res. 2014;76:2–10. doi: 10.1038/pr.2014.49. [DOI] [PubMed] [Google Scholar]
  • 10.Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol. 2018;11(1):1–10. doi: 10.1007/s12328-017-0813-5. [DOI] [PubMed] [Google Scholar]
  • 11.Alavi S, Mitchell JD, Cho JY, et al. Interpersonal gut microbiome variation drives susceptibility and resistance to cholera infection. Cell. 2020;181:1533–1546. doi: 10.1016/J.CELL.2020.05.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Santiago-Rodriguez TM, Hollister EB. Multi ’omic data integration: a review of concepts, considerations, and approaches. Semin Perinatol. 2021 doi: 10.1016/J.SEMPERI.2021.151456. [DOI] [PubMed] [Google Scholar]
  • 13.Das B, Nair GB. Homeostasis and dysbiosis of the gut microbiome in health and disease. J Biosci. 2019;44:1–8. doi: 10.1007/S12038-019-9926-Y/TABLES/1. [DOI] [PubMed] [Google Scholar]
  • 14.Sommer F, Anderson JM, Bharti R, Raes J, Rosenstiel P. The resilience of the intestinal microbiota influences health and disease. Nat Rev Microbiol. 2017;15(10):630–638. doi: 10.1038/nrmicro.2017.58. [DOI] [PubMed] [Google Scholar]
  • 15.Allam-Ndoul B, Castonguay-Paradis S, Veilleux A. Gut microbiota and intestinal trans-epithelial permeability. Int J Mol Sci. 2020;21:1–14. doi: 10.3390/IJMS21176402. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Paray BA, Albeshr MF, Jan AT, Rather IA. Leaky gut and autoimmunity: an intricate balance in individuals health and the diseased state. Int J Mol Sci. 2020;21:1–12. doi: 10.3390/IJMS21249770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Hall A, Tolonen A, Xavier R. Human genetic variation and the gut microbiome in disease. Nat Rev Genet. 2017;18:690–699. doi: 10.1038/NRG.2017.63. [DOI] [PubMed] [Google Scholar]
  • 18.Cheng S, Hu J, Wu X, et al. Altered gut microbiome in FUT2 loss-of-function mutants in support of personalized medicine for inflammatory bowel diseases. J Genet Genomics. 2021;48:771–780. doi: 10.1016/J.JGG.2021.08.003. [DOI] [PubMed] [Google Scholar]
  • 19.Moltzau Anderson J, Lipinski S, Sommer F, et al. NOD2 influences trajectories of intestinal microbiota recovery after antibiotic perturbation. Cell Mol Gastroenterol Hepatol. 2020;10:365–389. doi: 10.1016/J.JCMGH.2020.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Heianza Y, Sun D, Ma W, et al. Gut-microbiome-related LCT genotype and 2-year changes in body composition and fat distribution: the POUNDS Lost Trial. Int J Obes (Lond) 2018;42:1565–1573. doi: 10.1038/S41366-018-0046-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Kim H, Sitarik A, Woodcroft K, et al. Birth mode, breastfeeding, pet exposure, and antibiotic use: associations with the gut microbiome and sensitization in children. Curr Allergy Asthma Rep. 2019 doi: 10.1007/S11882-019-0851-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Arboleya S, Suárez M, Fernández N, et al. C-section and the neonatal gut microbiome acquisition: consequences for future Health. Ann Nutr Metab. 2018;73(3):17–23. doi: 10.1159/000490843. [DOI] [PubMed] [Google Scholar]
  • 23.Pannaraj P, Li F, Cerini C, et al. Association between breast milk bacterial communities and establishment and development of the infant gut microbiome. JAMA Pediatr. 2017;171:647–654. doi: 10.1001/JAMAPEDIATRICS.2017.0378. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Milani C, Duranti S, Bottacini F, et al. The first microbial colonizers of the human gut: composition, activities, and health implications of the infant gut microbiota. Microbiol Mol Biol Rev. 2017 doi: 10.1128/MMBR.00036-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, Gordon JI. Human gut microbiome viewed across age and geography. Nature. 2012;486(7402):222–227. doi: 10.1038/nature11053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Vaiserman A, Romanenko M, Piven L, et al. Differences in the gut Firmicutes to Bacteroidetes ratio across age groups in healthy Ukrainian population. BMC Microbiol. 2020;20:1–8. doi: 10.1186/S12866-020-01903-7/TABLES/4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Salazar N, Valdés-Varela L, González S, Gueimonde M, De Los Reyes-Gavilán CG. Nutrition and the gut microbiome in the elderly. Gut Microbes. 2017;8(2):82–97. doi: 10.1080/19490976.2016.1256525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hylander BL, Repasky EA. Temperature as a modulator of the gut microbiome: what are the implications and opportunities for thermal medicine? Int J Hyperthermia. 2019;36:83–89. doi: 10.1080/02656736.2019.1647356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Manor O, Dai CL, Kornilov SA, et al. Health and disease markers correlate with gut microbiome composition across thousands of people. Nat Commun. 2020 doi: 10.1038/S41467-020-18871-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Chen L, Wang D, Garmaeva S, et al. The long-term genetic stability and individual specificity of the human gut microbiome. Cell. 2021;184:2302–2315.e12. doi: 10.1016/J.CELL.2021.03.024. [DOI] [PubMed] [Google Scholar]
  • 31.Peace O, Rachakonda K, Kress M, et al. Respiratory and neurological disease across different ethnic groups is influenced by the microbiome. Microorganisms. 2021 doi: 10.3390/MICROORGANISMS9091965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Gupta V, Paul S, Dutta C. Geography, ethnicity or subsistence-specific variations in human microbiome composition and diversity. Front Microbiol. 2017 doi: 10.3389/FMICB.2017.01162. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Amato KR, Arrieta MC, Azad MB, et al. The human gut microbiome and health inequities. Proc Natl Acad Sci U S A. 2021 doi: 10.1073/pnas.2017947118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Shanahan F, Ghosh T, O’Toole P. The healthy microbiome-what is the definition of a healthy gut microbiome? Gastroenterology. 2021;160:483–494. doi: 10.1053/J.GASTRO.2020.09.057. [DOI] [PubMed] [Google Scholar]
  • 35.Keohane DM, Ghosh TS, Jeffery IB, Molloy MG, O’Toole PW, Shanahan F. Microbiome and health implications for ethnic minorities after enforced lifestyle changes. Nature Med. 2020;26(7):1089–1095. doi: 10.1038/s41591-020-0963-8. [DOI] [PubMed] [Google Scholar]
  • 36.Brooks AW, Priya S, Blekhman R, Bordenstein SR. Gut microbiota diversity across ethnicities in the United States. PLoS Biol. 2018;16:e2006842. doi: 10.1371/JOURNAL.PBIO.2006842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Bibbò S, Ianiro G, Giorgio V, et al. The role of diet on gut microbiota composition. Eur Rev Med Pharmacol Sci. 2016;20:4742–4749. [PubMed] [Google Scholar]
  • 38.Wang DD, Nguyen LH, Li Y, et al. The gut microbiome modulates the protective association between a Mediterranean diet and cardiometabolic disease risk. Nat Med. 2021;27:333–343. doi: 10.1038/S41591-020-01223-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bolte LA, Vich Vila A, Imhann F, et al. Original research: Long-term dietary patterns are associated with pro-inflammatory and anti-inflammatory features of the gut microbiome. Gut. 2021;70:1287. doi: 10.1136/GUTJNL-2020-322670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Savin Z, Kivity S, Yonath H, Yehuda S. Smoking and the intestinal microbiome. Arch Microbiol. 2018;200:677–684. doi: 10.1007/S00203-018-1506-2. [DOI] [PubMed] [Google Scholar]
  • 41.Engen PA, Green SJ, Voigt RM, Forsyth CB, Keshavarzian A. The gastrointestinal microbiome: alcohol effects on the composition of intestinal microbiota. Alcohol Res Curr Rev. 2015;37(2):223. [PMC free article] [PubMed] [Google Scholar]
  • 42.Mailing L, Allen J, Buford T, et al. Exercise and the gut microbiome: a review of the evidence, potential mechanisms, and implications for human health. Exerc Sport Sci Rev. 2019;47:75–85. doi: 10.1249/JES.0000000000000183. [DOI] [PubMed] [Google Scholar]
  • 43.Mahajan C, Khurana S, Kapoor I, et al. Characteristics of gut microbiome after traumatic brain injury. J Neurosurg Anesthesiol. 2021 doi: 10.1097/ANA.0000000000000789. [DOI] [PubMed] [Google Scholar]
  • 44.Aghakhani N. Relationship between mild traumatic brain injury and the gut microbiome: a scoping review. J Neurosci Res. 2021 doi: 10.1002/JNR.25004. [DOI] [PubMed] [Google Scholar]
  • 45.Kigerl KA, Hall JCE, Wang L, et al. Gut dysbiosis impairs recovery after spinal cord injury. J Exp Med. 2016;213:2603–2620. doi: 10.1084/jem.20151345. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Wallace DJ, Sayre NL, Patterson TT, et al. Spinal cord injury and the human microbiome: beyond the brain-gut axis. Neurosurg Focus. 2019 doi: 10.3171/2018.12.FOCUS18206. [DOI] [PubMed] [Google Scholar]
  • 47.Konturek PC, Brzozowski T, Konturek SJ. Stress and the gut: Pathophysiology, clinical consequences, diagnostic approach and treatment options. J Physiol Pharmacol. 2011;62:591–599. [PubMed] [Google Scholar]
  • 48.van de Wouw M, Boehme M, Lyte JM, et al. Short-chain fatty acids: microbial metabolites that alleviate stress-induced brain–gut axis alterations. J Physiol. 2018;596:4923–4944. doi: 10.1113/JP276431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Tsai YL, Lin TL, Chang CJ, et al. Probiotics, prebiotics and amelioration of diseases. J Biomed Sci. 2019 doi: 10.1186/S12929-018-0493-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Quigley EMM. Prebiotics and Probiotics in Digestive Health. Clin Gastroenterol Hepatol. 2019;17:333–344. doi: 10.1016/J.CGH.2018.09.028. [DOI] [PubMed] [Google Scholar]
  • 51.Gomaa EZ. Human gut microbiota/microbiome in health and diseases: a review. Antonie van Leeuwenhoek Int J Gen Mol Microbiol. 2020;113:2019–2040. doi: 10.1007/s10482-020-01474-7. [DOI] [PubMed] [Google Scholar]
  • 52.Ribeiro C, Silveira G, Cândido E, et al. Effects of antibiotic treatment on gut microbiota and how to overcome its negative impacts on human health. ACS Infect Dis. 2020;6:2544–2559. doi: 10.1021/ACSINFECDIS.0C00036. [DOI] [PubMed] [Google Scholar]
  • 53.Macke L, Schulz C, Koletzko L, Malfertheiner P. Systematic review: the effects of proton pump inhibitors on the microbiome of the digestive tract-evidence from next-generation sequencing studies. Aliment Pharmacol Ther. 2020;51:505–526. doi: 10.1111/APT.15604. [DOI] [PubMed] [Google Scholar]
  • 54.Tropini C, Moss EL, Merrill BD, Ng KM, Higginbottom SK, Casavant EP, Gonzalez CG, Fremin B, Bouley DM, Elias JE, Bhatt AS, Huang KC, Sonnenburg JL. Transient osmotic perturbation causes long-term alteration to the gut microbiota. Cell. 2018;173(7):1742–1754.e17. doi: 10.1016/j.cell.2018.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Zhang Q, Hu N. Effects of metformin on the gut microbiota in obesity and type 2 diabetes mellitus. Diabetes Metab Syndr Obes. 2020;13:5003–5014. doi: 10.2147/DMSO.S286430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Turksoy VA, Tutkun L, Gunduzoz M, et al. Changing levels of selenium and zinc in cadmium-exposed workers: probable association with the intensity of inflammation. Mol Biol Rep. 2019;46:5455–5464. doi: 10.1007/s11033-019-05001-4. [DOI] [PubMed] [Google Scholar]
  • 57.Li X, Brejnrod AD, Ernst M, et al. Heavy metal exposure causes changes in the metabolic health-associated gut microbiome and metabolites. Environ Int. 2019;126:454–467. doi: 10.1016/j.envint.2019.02.048. [DOI] [PubMed] [Google Scholar]
  • 58.Rueda-Ruzafa L, Cruz F, Roman P, Cardona D. Gut microbiota and neurological effects of glyphosate. Neurotoxicology. 2019;75:1–8. doi: 10.1016/J.NEURO.2019.08.006. [DOI] [PubMed] [Google Scholar]
  • 59.Petersen C, Round JL. Defining dysbiosis and its influence on host immunity and disease. Cell Microbiol. 2014;16:1024. doi: 10.1111/CMI.12308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Camilleri M. The leaky gut: mechanisms, measurement and clinical implications in humans. Gut. 2019;68:1516. doi: 10.1136/GUTJNL-2019-318427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kinashi Y, Hase K. Partners in leaky gut syndrome: intestinal dysbiosis and autoimmunity. Front Immunol. 2021 doi: 10.3389/FIMMU.2021.673708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Ghaisas S, Maher J, Kanthasamy A. Gut microbiome in health and disease: Linking the microbiome-gut-brain axis and environmental factors in the pathogenesis of systemic and neurodegenerative diseases. Pharmacol Ther. 2016;158:52–62. doi: 10.1016/j.pharmthera.2015.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Dabke K, Hendrick G, Devkota S. The gut microbiome and metabolic syndrome. J Clin Investig. 2019;129:4050–4057. doi: 10.1172/JCI129194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chen J, Wang A, Wang Q. Dysbiosis of the gut microbiome is a risk factor for osteoarthritis in older female adults: a case control study. BMC Bioinformatics. 2021;22:299. doi: 10.1186/s12859-021-04199-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Hu S, Png E, Gowans M, et al. Ectopic gut colonization: a metagenomic study of the oral and gut microbiome in Crohn’s disease. Gut Pathog. 2021 doi: 10.1186/s13099-021-00409-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Miquel S, Martín R, Lashermes A, et al. Anti-nociceptive effect of Faecalibacterium prausnitzii in non-inflammatory IBS-like models. Sci Rep. 2016 doi: 10.1038/srep19399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Boer CG, Radjabzadeh D, Medina-Gomez C, et al. Intestinal microbiome composition and its relation to joint pain and inflammation. Nat Commun. 2019;10:1–9. doi: 10.1038/s41467-019-12873-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Freidin MB, Stalteri MA, Wells PM, et al. An association between chronic widespread pain and the gut microbiome. Rheumatology. 2020 doi: 10.1093/rheumatology/keaa847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Minerbi A, Gonzalez E, Brereton NJB, et al. Altered microbiome composition in individuals with fibromyalgia. Pain. 2019;160:2589–2602. doi: 10.1097/j.pain.0000000000001640. [DOI] [PubMed] [Google Scholar]
  • 70.Seifert O, Baerwald C. Interaction of pain and chronic inflammation. Z Rheumatol. 2021;80:205–213. doi: 10.1007/S00393-020-00951-8. [DOI] [PubMed] [Google Scholar]
  • 71.Zhou WBS, Meng J, Zhang J. Does low grade systemic inflammation have a role in chronic pain? Front Mol Neurosci. 2021 doi: 10.3389/FNMOL.2021.785214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Schirmer M, Smeekens SP, Vlamakis H, et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell. 2016;167:1897. doi: 10.1016/J.CELL.2016.11.046. [DOI] [PubMed] [Google Scholar]
  • 73.Yeoh YK, Zuo T, Lui GCY, et al. Original research: Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut. 2021;70:698. doi: 10.1136/GUTJNL-2020-323020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Sethi V, Kurtom S, Tarique M, et al. Gut microbiota promotes tumor growth in mice by modulating immune response. Gastroenterology. 2018;155:33. doi: 10.1053/J.GASTRO.2018.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Stacy A, Andrade-Oliveira V, McCulloch JA, et al. Infection trains the host for microbiota-enhanced resistance to pathogens. Cell. 2021;184:615–627.e17. doi: 10.1016/J.CELL.2020.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Lee CJ, Sears CL, Maruthur N. Gut microbiome and its role in obesity and insulin resistance. Ann N Y Acad Sci. 2020;1461:37–52. doi: 10.1111/NYAS.14107. [DOI] [PubMed] [Google Scholar]
  • 77.Clark A, Mach N. Exercise-induced stress behavior, gut-microbiota-brain axis and diet: a systematic review for athletes. J Int Soc Sports Nutr. 2016 doi: 10.1186/S12970-016-0155-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Brevi A, Cogrossi LL, Grazia G, et al. Much more than IL-17A: cytokines of the IL-17 family between microbiota and cancer. Front Immunol. 2020 doi: 10.3389/FIMMU.2020.565470. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Parker A, Fonseca S, Carding SR. Gut microbes and metabolites as modulators of blood-brain barrier integrity and brain health. Gut Microbes. 2020;11:135. doi: 10.1080/19490976.2019.1638722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Haase S, Haghikia A, Wilck N, et al. Impacts of microbiome metabolites on immune regulation and autoimmunity. Immunology. 2018;154:230. doi: 10.1111/IMM.12933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Ohira H, Tsutsui W, Fujioka Y. Are short chain fatty acids in gut microbiota defensive players for inflammation and atherosclerosis? J Atheroscler Thromb. 2017;24:660. doi: 10.5551/JAT.RV17006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Venegas DP, de La Fuente MK, Landskron G, et al. Short chain fatty acids (SCFAs)mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol. 2019;10:277. doi: 10.3389/FIMMU.2019.00277/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Cortes-Altamirano JL, Olmos-Hernandez A, Jaime HB, et al. Review: 5-HT1, 5-HT2, 5-HT3 and 5-HT7 receptors and their role in the modulation of pain response in the central nervous system. Curr Neuropharmacol. 2018 doi: 10.2174/1570159X15666170911121027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Serafini RA, Pryce KD, Zachariou V. The mesolimbic dopamine system in chronic pain and associated affective comorbidities. Biol Psychiatry. 2020;87:64–73. doi: 10.1016/J.BIOPSYCH.2019.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.González-Arancibia C, Urrutia-Piñones J, Illanes-González J, et al. Do your gut microbes affect your brain dopamine? Psychopharmacology. 2019;236:1611–1622. doi: 10.1007/S00213-019-05265-5. [DOI] [PubMed] [Google Scholar]
  • 86.Pertovaara A. Noradrenergic pain modulation. Prog Neurobiol. 2006;80:53–83. doi: 10.1016/J.PNEUROBIO.2006.08.001. [DOI] [PubMed] [Google Scholar]
  • 87.Peek AL, Rebbeck T, Puts NA, et al. Brain GABA and glutamate levels across pain conditions: a systematic literature review and meta-analysis of 1H-MRS studies using the MRS-Q quality assessment tool. Neuroimage. 2020;210:116532. doi: 10.1016/J.NEUROIMAGE.2020.116532. [DOI] [PubMed] [Google Scholar]
  • 88.Zunhammer M, Schweizer LM, Witte V, et al. Combined glutamate and glutamine levels in pain-processing brain regions are associated with individual pain sensitivity. Pain. 2016;157:2248–2256. doi: 10.1097/J.PAIN.0000000000000634. [DOI] [PubMed] [Google Scholar]
  • 89.Hannibal KE, Bishop MD. Chronic stress, cortisol dysfunction, and pain: a psychoneuroendocrine rationale for stress management in pain rehabilitation. Phys Ther. 2014;94:1816–1825. doi: 10.2522/PTJ.20130597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Liang Y, Ma Y, Wang J, et al. Leptin contributes to neuropathic pain via Extrasynaptic NMDAR-nNOS activation. Mol Neurobiol. 2021;58:1185–1195. doi: 10.1007/S12035-020-02180-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Kinfe TM, Buchfelder M, Chaudhry SR, et al. Leptin and associated mediators of immunometabolic signaling: novel molecular outcome measures for neurostimulation to treat chronic pain. Int J Mol Sci. 2019 doi: 10.3390/IJMS20194737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Lippi G, Dagostino C, Buonocore R, et al. The serum concentrations of leptin and MCP-1 independently predict low back pain duration. Clin Chem Lab Med. 2017;55:1368–1374. doi: 10.1515/CCLM-2016-0942. [DOI] [PubMed] [Google Scholar]
  • 93.Lach G, Schellekens H, Dinan TG, Cryan JF. Anxiety, depression, and the microbiome: a role for gut peptides. Neurotherapeutics. 2018;15:36. doi: 10.1007/S13311-017-0585-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Helde-Frankling M, Björkhem-Bergman L. Vitamin D in pain management. Int J Mol Sci. 2017 doi: 10.3390/ijms18102170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Yong WC, Sanguankeo A, Upala S. Effect of vitamin D supplementation in chronic widespread pain: a systematic review and meta-analysis. Clin Rheumatol. 2017;36:2825–2833. doi: 10.1007/s10067-017-3754-y. [DOI] [PubMed] [Google Scholar]
  • 96.Ang CD, Alviar MJM, Dans AL, et al. Vitamin B for treating peripheral neuropathy. Cochrane Database Syst Rev. 2008 doi: 10.1002/14651858.CD004573.PUB3. [DOI] [PubMed] [Google Scholar]
  • 97.Buesing S, Costa M, Schilling JM, Moeller-Bertram T. Vitamin B12 as a treatment for pain. Pain Phys. 2019;22(1):E45. doi: 10.36076/ppj/2019.22.E45. [DOI] [PubMed] [Google Scholar]
  • 98.Clarke G, Stilling RM, Kennedy PJ, et al. Minireview: gut microbiota: the neglected endocrine organ. Mol Endocrinol. 2014;28:1221–1238. doi: 10.1210/me.2014-1108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Silva YP, Bernardi A, Frozza RL. The Role of Short-Chain Fatty Acids From Gut Microbiota in Gut-Brain Communication. Front Endocrinol (Lausanne) 2020;11:25. doi: 10.3389/FENDO.2020.00025/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Xue R, Zhang H, Pan J, et al. Peripheral dopamine controlled by gut microbes inhibits invariant natural killer T cell-mediated hepatitis. Front Immunol. 2018;9:2398. doi: 10.3389/FIMMU.2018.02398/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Vidal PM, Pacheco R. The cross-talk between the dopaminergic and the immune system involved in schizophrenia. Front Pharmacol. 2020;11:394. doi: 10.3389/FPHAR.2020.00394/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Valles-Colomer M, Falony G, Darzi Y, et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat Microbiol. 2019;4:623–632. doi: 10.1038/S41564-018-0337-X. [DOI] [PubMed] [Google Scholar]
  • 103.Ziółkowska B. The role of mesostriatal dopamine system and corticostriatal glutamatergic transmission in chronic pain. Brain Sci. 2021 doi: 10.3390/BRAINSCI11101311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Berger M, Gray JA, Roth BL. The expanded biology of serotonin. Annu Rev Med. 2009;60:355–366. doi: 10.1146/ANNUREV.MED.60.042307.110802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Daut RA, Fonken LK. Circadian regulation of depression: a role for serotonin. Front Neuroendocrinol. 2019 doi: 10.1016/J.YFRNE.2019.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Meneses A, Liy-Salmeron G. Serotonin and emotion, learning and memory. Rev Neurosci. 2012;23:543–553. doi: 10.1515/REVNEURO-2012-0060. [DOI] [PubMed] [Google Scholar]
  • 107.Yano JM, Yu K, Donaldson GP, et al. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell. 2015;161:264–276. doi: 10.1016/J.CELL.2015.02.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Malinova TS, Dijkstra CD, de Vries HE. Serotonin: a mediator of the gut–brain axis in multiple sclerosis. Mult Scler. 2018;24:1144. doi: 10.1177/1352458517739975. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Stasi C, Sadalla S, Milani S. The relationship between the serotonin metabolism, gut-microbiota and the gut-brain axis. Curr Drug Metab. 2019;20:646–655. doi: 10.2174/1389200220666190725115503. [DOI] [PubMed] [Google Scholar]
  • 110.Israelyan N, Margolis KG. Serotonin as a link between the gut-brain-microbiome axis in autism spectrum disorders. Pharmacol Res. 2019;140:115. doi: 10.1016/J.PHRS.2018.12.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Layunta E, Buey B, Mesonero JE, Latorre E. Crosstalk between intestinal serotonergic system and pattern recognition receptors on the microbiota–gut–brain axis. Front Endocrinol (Lausanne) 2021;12:1379. doi: 10.3389/FENDO.2021.748254/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Cryan JF, O’riordan KJ, Cowan CSM, et al. The microbiota-gut-brain axis. Physiol Rev. 2019;99:1877–2013. doi: 10.1152/PHYSREV.00018.2018/ASSET/IMAGES/LARGE/Z9J0041929160006.JPEG. [DOI] [PubMed] [Google Scholar]
  • 113.Gao J, Xu K, Liu H, et al. Impact of the gut microbiota on intestinal immunity mediated by tryptophan metabolism. Front Cell Infect Microbiol. 2018;8:13. doi: 10.3389/FCIMB.2018.00013/BIBTEX. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.O’Mahony SM, Clarke G, Borre YE, et al. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav Brain Res. 2015;277:32–48. doi: 10.1016/j.bbr.2014.07.027. [DOI] [PubMed] [Google Scholar]
  • 115.Russo R, Cristiano C, Avagliano C, et al. Gut-brain axis: role of lipids in the regulation of inflammation, pain and CNS diseases. Curr Med Chem. 2018;25:3930–3952. doi: 10.2174/0929867324666170216113756. [DOI] [PubMed] [Google Scholar]
  • 116.Raskov H, Burcharth J, Pommergaard HC, Rosenberg J. Irritable bowel syndrome, the microbiota and the gut-brain axis. Gut Microbes. 2016;7:365. doi: 10.1080/19490976.2016.1218585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Liu J, Xu F, Nie Z, Shao L. Gut microbiota approach—a new strategy to treat Parkinson’s disease. Front Cell Infect Microbiol. 2020 doi: 10.3389/FCIMB.2020.570658. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Barandouzi ZA, Lee J, del Carmen Rosas M, Chen J, Henderson WA, Starkweather AR, Cong XS. Associations of neurotransmitters and the gut microbiome with emotional distress in mixed type of irritable bowel syndrome. Sci Rep. 2022;12(1):1–11. doi: 10.1038/S41598-022-05756-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Schnabel A, Weibel S, Reichl SU, et al. Efficacy and adverse events of selective serotonin noradrenaline reuptake inhibitors in the management of postoperative pain: a systematic review and meta-analysis. J Clin Anesth. 2021 doi: 10.1016/J.JCLINANE.2021.110451. [DOI] [PubMed] [Google Scholar]
  • 120.O’Neill E, Harkin A. Targeting the noradrenergic system for anti-inflammatory and neuroprotective effects: implications for Parkinson’s disease. Neural Regen Res. 2018;13(8):1332. doi: 10.4103/1673-5374.235219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Sharma D, Farrar JD. Adrenergic regulation of immune cell function and inflammation. Semin Immunopathol. 2020;42:1. doi: 10.1007/S00281-020-00829-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Pongratz G, Straub RH. The sympathetic nervous response in inflammation. Arthritis Res Ther. 2014;16:1–12. doi: 10.1186/S13075-014-0504-2/FIGURES/4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Leach S, Suzuki K. Adrenergic signaling in circadian control of immunity. Front Immunol. 2020 doi: 10.3389/FIMMU.2020.01235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Sherwani ZA, Khalil R, Nur-e-Alam M, et al. Structure-based virtual screening to identify negative allosteric modulators of NMDA. Med Chem. 2022 doi: 10.2174/1573406418666220304224150. [DOI] [PubMed] [Google Scholar]
  • 125.Zhao H, Mao X, Zhu C, et al. GABAergic system dysfunction in autism spectrum disorders. Front Cell Dev Biol. 2022 doi: 10.3389/FCELL.2021.781327. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Auteri M, Zizzo MG, Serio R. GABA and GABA receptors in the gastrointestinal tract: from motility to inflammation. Pharmacol Res. 2015;93:11–21. doi: 10.1016/J.PHRS.2014.12.001. [DOI] [PubMed] [Google Scholar]
  • 127.Dinan TG, Cryan JF. The microbiome-gut-brain axis in health and disease. Gastroenterol Clin North Am. 2017;46:77–89. doi: 10.1016/J.GTC.2016.09.007. [DOI] [PubMed] [Google Scholar]
  • 128.Strandwitz P, Kim KH, Terekhova D, et al. GABA-modulating bacteria of the human gut microbiota. Nat Microbiol. 2019;4:396–403. doi: 10.1038/S41564-018-0307-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Stephens MAC, Wand G. Stress and the HPA Axis: role of glucocorticoids in alcohol dependence. Alcohol Res. 2012;34:468. [PMC free article] [PubMed] [Google Scholar]
  • 130.Lee DY, Kim E, Choi MH. Technical and clinical aspects of cortisol as a biochemical marker of chronic stress. BMB Rep. 2015;48:209. doi: 10.5483/BMBREP.2015.48.4.275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Villafañe JH, Pedersini P, Bertozzi L, et al. Exploring the relationship between chronic pain and cortisol levels in subjects with osteoarthritis: results from a systematic review of the literature. Osteoarthritis Cartilage. 2020;28:572–580. doi: 10.1016/J.JOCA.2020.02.836. [DOI] [PubMed] [Google Scholar]
  • 132.Dinan TG, Cryan JF. Regulation of the stress response by the gut microbiota: implications for psychoneuroendocrinology. Psychoneuroendocrinology. 2012;37:1369–1378. doi: 10.1016/J.PSYNEUEN.2012.03.007. [DOI] [PubMed] [Google Scholar]
  • 133.Wu B, Liu Y, Liu F, et al. The antinociceptive effects and molecular mechanisms of ghrelin(1–7)-NH 2 at the supraspinal level in acute pain in mice. Brain Res Bull. 2019;146:112–123. doi: 10.1016/J.BRAINRESBULL.2018.12.016. [DOI] [PubMed] [Google Scholar]
  • 134.Peng Z, Zha L, Yang M, et al. Effects of ghrelin on pGSK-3β and β-catenin expression when protects against neuropathic pain behavior in rats challenged with chronic constriction injury. Sci Rep. 2019 doi: 10.1038/S41598-019-51140-W. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Azizzadeh F, Mahmoodi J, Sadigh-Eteghad S, Farajdokht F, Mohaddes G. Ghrelin exerts analgesic effects through modulation of IL-10 and TGF-β levels in a rat model of inflammatory pain. Iran Biomed J. 2017;21(2):114. doi: 10.18869/acadpub.ibj.21.2.114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Xu Q, Jiang M, Gu S, et al. Metabolomics changes in brain-gut axis after unpredictable chronic mild stress. Psychopharmacology. 2022;239:729–743. doi: 10.1007/S00213-021-05958-W. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Han H, Zhou W. Leptin and Its derivatives: a potential target for autoimmune diseases. Curr Drug Targets. 2019;20:1563–1571. doi: 10.2174/1389450120666190729120557. [DOI] [PubMed] [Google Scholar]
  • 138.ter Horst R, van den Munckhof ICL, Schraa K, et al. Sex-specific regulation of inflammation and metabolic syndrome in obesity. Arterioscler Thromb Vasc Biol. 2020;40:1787–1800. doi: 10.1161/ATVBAHA.120.314508. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Song X, Zhong L, Lyu N, et al. Inulin can alleviate metabolism disorders in ob/ob mice by partially restoring leptin-related pathways mediated by gut microbiota. Genomics Proteomics Bioinform. 2019;17:64–75. doi: 10.1016/J.GPB.2019.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Heiss CN, Olofsson LE. Gut microbiota-dependent modulation of energy metabolism. J Innate Immun. 2018;10:163–171. doi: 10.1159/000481519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Rowland I, Gibson G, Heinken A, et al. Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr. 2018;57:1. doi: 10.1007/S00394-017-1445-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Rudzki L, Stone TW, Maes M, et al. Gut microbiota-derived vitamins - underrated powers of a multipotent ally in psychiatric health and disease. Prog Neuropsychopharmacol Biol Psychiatry. 2021 doi: 10.1016/J.PNPBP.2020.110240. [DOI] [PubMed] [Google Scholar]
  • 143.Fakhoury HMA, Kvietys PR, AlKattan W, et al. Vitamin D and intestinal homeostasis: barrier, microbiota, and immune modulation. J Steroid Biochem Mol Biol. 2020 doi: 10.1016/J.JSBMB.2020.105663. [DOI] [PubMed] [Google Scholar]
  • 144.Zadro J, Shirley D, Ferreira M, et al. Mapping the association between vitamin d and low back pain: a systematic review and meta-analysis of observational studies. Pain Phys. 2017;20:611–640. doi: 10.36076/ppj/2017.7.611. [DOI] [PubMed] [Google Scholar]
  • 145.Wong CC, Qian Y, Yu J. Interplay between epigenetics and metabolism in oncogenesis: mechanisms and therapeutic approaches. Oncogene. 2017;36(24):3359–3374. doi: 10.1038/onc.2016.485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Zhang D, Tang Z, Huang H, et al. Metabolic regulation of gene expression by histone lactylation. Nature. 2019;574:575–580. doi: 10.1038/S41586-019-1678-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Boison D. The biochemistry and epigenetics of epilepsy: focus on adenosine and glycine. Front Mol Neurosci. 2016 doi: 10.3389/FNMOL.2016.00026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Kim H, Park YJ. Links between serine biosynthesis pathway and epigenetics in cancer metabolism. Clin Nutr Res. 2018;7:153. doi: 10.7762/CNR.2018.7.3.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Wang Z, Yip LY, Lee JHJ, et al. Methionine is a metabolic dependency of tumor-initiating cells. Nat Med. 2019 doi: 10.1038/S41591-019-0423-5. [DOI] [PubMed] [Google Scholar]
  • 150.Wu B, Qiu J, Zhao TV, et al. Succinyl-CoA ligase deficiency in pro-inflammatory and tissue-invasive T cells. Cell Metab. 2020;32:967–980.e5. doi: 10.1016/j.cmet.2020.10.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Certo M, Tsai CH, Pucino V, Ho PC, Mauro C. Lactate modulation of immune responses in inflammatory versus tumour microenvironments. Nat Rev Immunol. 2021;21(3):151–161. doi: 10.1038/s41577-020-0406-2. [DOI] [PubMed] [Google Scholar]
  • 152.Krautkramer KA, Dhillon RS, Denu JM, Carey HV. Metabolic programming of the epigenome: host and gut microbial metabolite interactions with host chromatin. Trans Res. 2017;189:30. doi: 10.1016/J.TRSL.2017.08.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Amaral FA, Sachs D, Costa VV, et al. Commensal microbiota is fundamental for the development of inflammatory pain. Proc Natl Acad Sci U S A. 2008;105:2193–2197. doi: 10.1073/pnas.0711891105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Ma Y, Liu S, Shu H, et al. Resveratrol alleviates temporomandibular joint inflammatory pain by recovering disturbed gut microbiota. Brain Behav Immun. 2020;87:455–464. doi: 10.1016/J.BBI.2020.01.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Chen P, Wang C, Ren Y, na, , et al. Alterations in the gut microbiota and metabolite profiles in the context of neuropathic pain. Mol Brain. 2021 doi: 10.1186/S13041-021-00765-Y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Ding W, You Z, Chen Q, et al. Gut microbiota influences neuropathic pain through modulating proinflammatory and anti-inflammatory t cells. Anesth Analg. 2021;132:1146–1155. doi: 10.1213/ane.0000000000005155. [DOI] [PubMed] [Google Scholar]
  • 157.Ellis RJ, Heaton RK, Gianella S, et al. Reduced gut microbiome diversity in people with HIV who have distal neuropathic pain. J Pain. 2021 doi: 10.1016/J.JPAIN.2021.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Clos-Garcia M, Andrés-Marin N, Fernández-Eulate G, et al. Gut microbiome and serum metabolome analyses identify molecular biomarkers and altered glutamate metabolism in fibromyalgia. EBioMedicine. 2019;46:499–511. doi: 10.1016/j.ebiom.2019.07.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Shiro Y, Arai Y-C, Ikemoto T, et al. Correlation between gut microbiome composition and acute pain perception in young healthy male subjects. Pain Med. 2020;00:1–10. doi: 10.1093/pm/pnaa401. [DOI] [PubMed] [Google Scholar]
  • 160.Shiro Y, Arai YC, Ikemoto T, Hayashi K. Stool consistency is significantly associated with pain perception. PLoS ONE. 2017;12:2–10. doi: 10.1371/journal.pone.0182859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Vandeputte D, Falony G, Vieira-Silva S, et al. Original article: Stool consistency is strongly associated with gut microbiota richness and composition, enterotypes and bacterial growth rates. Gut. 2016;65:57. doi: 10.1136/GUTJNL-2015-309618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Akbarali HI, Dewey WL. The gut-brain interaction in opioid tolerance. Curr Opin Pharmacol. 2017;37:126. doi: 10.1016/J.COPH.2017.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Guido G, Ausenda G, Iascone V, Chisari E. Gut permeability and osteoarthritis, towards a mechanistic understanding of the pathogenesis: a systematic review. Ann Med. 2021;53:2380. doi: 10.1080/07853890.2021.2014557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Chisari E, Wouthuyzen-Bakker M, Friedrich AW, Parvizi J. The relation between the gut microbiome and osteoarthritis: A systematic review of literature. PLoS ONE. 2021;16:e0261353. doi: 10.1371/JOURNAL.PONE.0261353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Sánchez Romero EA, Meléndez Oliva E, Alonso Pérez JL, et al. Relationship between the gut microbiome and osteoarthritis pain: review of the literature. Nutrients. 2021 doi: 10.3390/nu13030716. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Boer CG, Radjabzadeh D, Medina-Gomez C, et al. Intestinal microbiome composition and its relation to joint pain and inflammation. Nat Commun. 2019;10:4881. doi: 10.1038/s41467-019-12873-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Wei J, Zhang C, Zhang Y, et al. Association between gut microbiota and symptomatic hand osteoarthritis: data from the xiangya osteoarthritis study. Arthritis Rheumatol. 2021;73:1656–1662. doi: 10.1002/ART.41729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Coulson S, Butt H, Vecchio P, et al. Green-lipped mussel extract (Perna canaliculus) and glucosamine sulphate in patients with knee osteoarthritis: therapeutic efficacy and effects on gastrointestinal microbiota profiles. Inflammopharmacology. 2013;21:79–90. doi: 10.1007/S10787-012-0146-4/TABLES/6. [DOI] [PubMed] [Google Scholar]
  • 169.Dekker Nitert M, Mousa A, Barrett HL, et al. Altered gut microbiota composition is associated with back pain in overweight and obese individuals. Front Endocrinol (Lausanne) 2020;11:1–10. doi: 10.3389/fendo.2020.00605. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from European Spine Journal are provided here courtesy of Nature Publishing Group

RESOURCES