Back to Journals » Journal of Inflammation Research » Volume 17

Unveiling the Gut-Disc Axis: How Microbiome Dysbiosis Accelerates Intervertebral Disc Degeneration

Authors Sun Y , Li Z, Duan J, Liu E, Sun F, Yang L, Chen L, Yang S

Received 20 July 2024

Accepted for publication 26 October 2024

Published 5 November 2024 Volume 2024:17 Pages 8271—8280

DOI https://doi.org/10.2147/JIR.S487936

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Dr Tara Strutt



Yu Sun,1 Zhaoyong Li,1 Jiahao Duan,1 Enxu Liu,1 Fei Sun,1 Lei Yang,2 Long Chen,2 Shaofeng Yang2

1Graduate School of Hunan University of Chinese Medicine, Changsha, Hunan, 410208, People’s Republic of China; 2Department of Orthopedics, the First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, 410007, People’s Republic of China

Correspondence: Shaofeng Yang, Department of Orthopedics, the First Affiliated Hospital of Hunan University of Chinese Medicine, No. 300 Xue-Shi Road, Yuelu District, Changsha, 410007, Hunan, People’s Republic of China, Tel +86 18391076233, Email [email protected]

Abstract: The gut microbiome (GM), often referred to as the second genome of the human body, plays a crucial role in various metabolic processes and mediates the development of numerous diseases. Intervertebral disc degeneration (IDD) is an age-related degenerative spinal disease characterized by the loss of disc height, hydration, and integrity, leading to pain and reduced mobility. Although the pathogenesis of IDD is not fully understood, recent studies suggest that dysbiosis of the gut microbiome may accelerate the progression of IDD through multiple mechanisms. This article begins by discussing the potential relationship between GM dysbiosis and human diseases, followed by a comprehensive review of the regulatory mechanisms of GM in skeletal diseases within the gut-disc axis framework. Furthermore, it explores three potential pathways through which GM dysbiosis may mediate the development of IDD: immunomodulation, bacterial translocation and colonization, and the decomposition and absorption of intestinal metabolites. These pathways can disrupt disc cell homeostasis and promote degenerative changes. Finally, this paper summarizes for the first time the potential therapeutic approaches for delaying IDD by targeting the gut-disc axis, providing new insights into the pathogenesis and regenerative repair strategies for IDD.

Keywords: intervertebral disc degeneration, microbiome, inflammation, immunity, metabolism

Intervertebral disc degeneration (IDD) is a complex pathological process involving multiple factors, including genetics, obesity, age, and lifestyle. It is primarily characterized by the loss of water content in the disc tissue, reduced elasticity, decreased height, and diminished ability to respond to mechanical loads.1,2

The human gut microbiome (GM) is a dynamic and complex ecosystem composed of countless microorganisms interacting with the human host.3,4 The GM, being the largest micro-ecosystem in the human body, plays a crucial role in maintaining normal physiological functions and metabolic balance. Dysbiosis of the GM, characterized by the overgrowth of pathogenic bacteria, is a micro-ecological imbalance associated with various diseases, including neurological disorders, cancer, and autoimmune diseases.5 However, the potential influence of this micro-ecological imbalance on IDD is still unclear. Therefore, this review provides a comprehensive narrative summary of the mechanisms by which GM regulates human diseases and explores potential ways in which GM influences IDD from various perspectives. Additionally, it provides the first summary of the latest research on targeting the gut-disc axis for the prevention and treatment of IDD, offering valuable insights into the association between spinal degenerative diseases and GM.

Overview of GM and Human Diseases

The gut microbiome (GM) plays a crucial role in maintaining host health through multiple pathways, including nutrient absorption, energy metabolism, and immune regulation. When the balance of GM becomes disrupted, both its structure and function become disordered, leading to the accumulation of dysregulated metabolic products that can influence the development of various diseases.6,7 Recent advancements in molecular biology, genomics, and large-scale parallel sequencing technologies have expanded our understanding of GM dysbiosis and its impacts. These imbalances not only disturb the intestinal environment but also mediate the progression of diseases across multiple systems, including the nervous, cardiovascular, and endocrine systems (as illustrated in Figure 1).

Figure 1 Overview of GM and Human Diseases. GM influences the development of neurological, cardiovascular, and endocrine diseases through multiple pathways.

In cardiovascular diseases (CVD), for instance, GM dysbiosis contributes to conditions like hypertension and atherosclerosis (ACVD) by influencing inflammation and vascular function through its metabolites. Hypertensive patients exhibit reduced GM diversity and abundance, with an increased Firmicutes/Bacteroidetes ratio.8 Studies have shown that they have significantly lower abundances of beneficial groups like Roseburia and Faecalibacterium, while pathogenic groups such as Enterobacteriaceae and Streptococcus are elevated.9,10 In ACVD, patients also demonstrate increased levels of Collinsella and Intestinimonas, contributing to disease progression.11 Similarly, in nervous system diseases like Parkinson’s disease (PD) and Alzheimer’s disease (AD), alterations in GM composition are closely linked to disease progression. In PD, increased abundances of Ruminococcaceae and Clostridium XIVa correlate with motor symptoms, while cognitive impairment is associated with changes in Butyricicoccus and Clostridium XIVa.12,13 In AD patients, the abundances of anti-inflammatory bacteria such as Eubacterium rectale, Bacteroides fragilis, and Faecalibacterium are significantly decreased, while pro-inflammatory genera like Escherichia and Shigella are increased.14,15 Additionally, AD patients show a lower abundance of Firmicutes and Actinobacteria, with a rise in Bacteroidetes.16 Endocrine system diseases, including obesity and type 2 diabetes mellitus (T2DM), are also characterized by GM dysbiosis. Obesity is marked by decreased levels of Akkermansia muciniphila, Faecalibacterium prausnitzii, and Bacteroides, while the abundance of Firmicutes is significantly increased.17,18 In T2DM, patients exhibit reduced gut microbiome diversity and an elevated Firmicutes/Bacteroidetes ratio, along with higher levels of Proteobacteria compared to healthy individuals.19,20 Understanding these GM shifts provides insights into the mechanisms underlying these multi-system diseases and presents potential avenues for novel therapeutic strategies.

Overview of GM and Bone Diseases

Under normal circumstances, a balanced GM abundance ratio efficiently regulates human metabolism and immune status. Recently, more scholars have proposed the microbiota-gut-bone axis concept to elucidate the complex relationship between gut microbiota and bone diseases.21,22 Building on this, Rajasekaran et al comprehensively evaluated the microbial composition of intervertebral discs (IVDs) and reported differences between the microbiota of normal and degenerated IVDs.23 This led to the gut-disc axis concept, indicating that GM may play a crucial regulatory role in IDD.24 This section presents an overview of the gut-bone axis and gut-disc axis concepts. Furthermore, it reviews the potential relationship between GM and bone and intervertebral disc tissue, providing insights into GM’s role in bone diseases.”

Gut-Bone Axis

Bone tissue, as a metabolically active organ of the human body, undergoes bone matrix formation, mineralization, and resorption throughout the entire bone remodeling process.25,26 This process is tightly regulated by the interaction between osteoblasts and osteoclasts, through direct cell contact or communication via secreted proteins, to regulate cell survival, differentiation, and apoptosis, which is crucial for maintaining bone homeostasis.27 The development of osteoporosis and osteoproliferative diseases involves immune-inflammatory regulatory mechanisms.28,29 The gut microbiome (GM) appears to play a role in this context, as it is thought to influence immunity and inflammation—key components of osteoimmunology. For instance, the GM is hypothesized to regulate cytokine secretion during metabolic processes, potentially through the production of short-chain fatty acids and other metabolites. This regulation may stimulate inflammation-mediated bone formation, although the direct association between GM-related cytokine release and disc degeneration remains to be fully established.30,31 It has been proposed that GM may secrete neurotransmitters that could activate the immune system, potentially leading to increased circulating osteoclast cytokines via T-cell-dependent mechanisms.32 However, it is important to note that these connections are still under investigation and may not indicate a causal relationship. Additionally, several gut hormones are involved in bone remodeling. After nutrient intake through food, intestinal endocrine cells secrete gut hormones that regulate glucose homeostasis and participate in energy requirements during bone remodeling.33 For example, gastric inhibitory polypeptide (GIP) and glucagon-like peptide 1 (GLP-1), which are primarily secreted by enteroendocrine cells, are thought to stimulate insulin secretion, potentially promoting insulin-mediated bone formation.34 Preclinical studies indicate that GIP receptor knockout mice exhibit significant decreases in bone mineral density (BMD) and bone strength, along with reductions in markers of bone formation such as alkaline phosphatase and osteocalcin.35 In summary, while the gut microbiome may influence the development of bone diseases through mechanisms such as nutrient absorption, inflammation, and immune regulation, the precise nature of these interactions, particularly their causal relationships, requires further investigation. A more nuanced understanding of the gut-bone axis may provide insights into potential strategies for preventing and treating bone diseases.

The Gut-Disc Axis

IVDs are avascular structures made of fibrocartilage, comprising three components: the central gelatinous nucleus pulposus, the outer fibrous annulus, and the upper and lower cartilaginous endplates. This structure separates vertebral bodies, transmits loads, and provides flexibility and effective stress support to the spine.36,37 Normally, IVDs are regarded as immune-privileged tissues lacking immune cells. The cells within, including CEP, NP, and AF, have been demonstrated to exhibit classical expression and markers of immune cells.38 However, the hypoxic environment and loss of immune barrier function in IVDs create conditions for anaerobic bacteria growth in degenerated IVDs. These bacteria produce large amounts of inflammatory factors, recruiting more inflammatory cells such as T cells, B cells, dendritic cells, and macrophages.39 This results in degenerated IVDs becoming ideal environments for microbial growth, proliferation, and metabolic product accumulation. Here, we explore three potential mechanisms through which gut microbiota dysbiosis accelerates IDD.

Immunomodulation

As mentioned above, the development of IDD have been proven to be associated with the abnormal induction of pro-inflammatory factor secretion by IVD cells and immune cells. These molecules trigger pathogenic and inflammatory responses in IVDs, potentially activating apoptosis, senescence, and autophagy.40,41 GM and the human immune system interact with each other. The types, abundance, and metabolites of GM regulate immune homeostasis and influence many immune-related diseases.42 Research has shown that in germ-free mouse models, the absence of GM results in decreased immune signaling, an imbalanced T(H)1/T(H)2 ratio, and systemic T cell deficiencies compared to mice colonized with the gut bacterium Bacteroides fragilis.43 Additionally, as a defense mechanism, intestinal epithelial cells produce a mucosal barrier that separates the microbiota from host immune cells, thereby reducing intestinal mucosal permeability.44 However, GM dysbiosis weakens the epithelial barrier, increasing permeability and contact between GM and the intestinal mucosal immune barrier, leading to an imbalance of potential pathogens.45 This excessive contact leads to an influx of activated immune cells and inflammatory cytokines, which migrate and accumulate near the IVDs via the bloodstream, thereby regulating bone metabolism and inducing the occurrence and development of IDD.39 Similarly, immune cells stimulate IVD cells to produce neurogenic factors such as neurotrophin-3 (NT-3), nerve growth factor (NGF), and brain-derived neurotrophic factor (BDNF), inducing the formation of nociceptive nerve fibers in the dorsal root ganglion (DRG).46 On the other hand, the persistent inflammatory environment in the degenerating IVD increases the expression of pain-related cation channels in the DRG, mediated by immune responses.47 Both are common causes of IDD-related low back pain. Therefore, GM dysbiosis may influence IDD development by affecting immune function in three proposed ways. Firstly, GM dysbiosis increases epithelial barrier permeability, allowing activated immune cells to migrate and accumulate near the IVDs via the bloodstream, regulating bone metabolism and inducing IDD. Secondly, the migration of immune cells into the IVDs stimulates IVD cells to produce neurogenic factors, leading to the emergence of nociceptive nerve fibers. Finally, the chronic inflammatory environment in IVDs increases the expression of pain-related cation channels, mediated by immune cells, contributing to IDD-associated low back pain.

Bacterial Translocation and Colonization

Recent studies have shown that inflammatory infiltration resulting from GM dysbiosis is a crucial factor in triggering bone and joint diseases such as osteoporosis and rheumatoid arthritis.48,49 Therefore, we hypothesize that chronic inflammation caused by GM imbalance may be an important factor in the occurrence and development of IDD. Below, we focus on the potential links among GM, inflammation, and IDD, while emphasizing the possible mechanism mediated by bacterial translocation and colonization.

As stated above, the protective effect of the blood-disc barrier renders healthy IVDs as immune-privileged tissues, effectively preventing the influence of other potential inflammations. However, this also impedes immune surveillance within the IVDs, preventing immune cells from promptly responding to microenvironment changes.40 Therefore, we propose that the lack of immune surveillance in the IVDs is a precondition for inducing IDD, and the inflammatory response triggered by anaerobic bacterial invasion within the IVDs is a critical factor in IDD. The interaction between these two factors jointly influences the occurrence and development of IDD.

Translocation and colonization of bacteria might mediate the development of IDD in three ways. On the one hand, chronic intestinal inflammatory changes caused by GM dysbiosis can increase intestinal permeability, promoting bacteria to penetrate the epithelial barrier and enter the bloodstream.50,51 Although most translocated bacteria are eradicated by the immune system, a few can survive the immune response, reaching the vicinity of the IVDs through the bloodstream, colonizing, and releasing pro-inflammatory factors that stimulate vascular proliferation into the IVDs, thereby disrupting their anaerobic environment. On the other hand, abnormal mechanical stress and sustained axial loading can also cause irreversible microdamage to the IVDs.52 This creates an ideal environment for bacterial growth and proliferation. Bacteria evading immune responses via the gut can enter the IVDs, with their pro-inflammatory factors and harmful metabolites further accelerating IDD progression. Lastly, bacteria entering through damaged IVDs can stimulate IVD cells to secrete inflammatory factors, affecting the balance of cellular synthesis and catabolism, leading to a cascade of reactions that promote chemokine production, ultimately accelerating collagen and proteoglycan inactivation within the IVDs and inducing Extracellular matrix (ECM) degradation.46

Decomposition and Absorption of Intestinal Metabolites

Under physiological conditions, intestinal epithelial mucus forms a robust physical barrier preventing pathogens from invading the intestine. Short-chain fatty acids (SCFAs), also known as volatile fatty acids, are organic fatty acids produced by the fermentation of carbohydrates by GM, with various physiological functions including energy provision, intestinal mucosal protection, metabolic regulation, and immune modulation.53 The colon is the primary site for SCFA production, mainly from the fermentation of undigested carbohydrates by anaerobic bacteria. Studies have demonstrated that SCFAs are essential metabolites produced by GM.54,55 The process of IDD is accompanied by vertebral bone remodeling, including endplate thickening and osteophyte formation. Similarly, IVD calcification is associated with the metabolic imbalance of osteoblasts and osteoclasts. Research shows that SCFAs promote the differentiation of immature CD4+ cells into regulatory T cells (Tregs), which preferentially reside on the bone endosteal surface.56 Furthermore, calcium deposition within IVDs and the expression of the extracellular calcium-sensing receptor (CaSR) are closely related to G-protein-coupled receptors in degenerated IVDs. SCFAs can inhibit bone resorption or osteoclast formation by activating G-protein-coupled receptors (GPCR) or inhibiting histone deacetylases (HDAC).57 This suggests that SCFAs derived from the gut may diffuse into the IVDs and accelerate the processes of calcification and IDD. Therefore, we believe SCFAs, key metabolites from GM, may play a crucial role in mediating IDD by affecting bone metabolic balance, inducing IVD calcification, and vertebral bone remodeling.

In conclusion, GM dysbiosis mediated by the gut-disc axis can accelerate IDD progression through immune regulation, bacterial translocation and colonization, and intestinal metabolite breakdown and absorption. This provides a reference for the prevention and treatment of IDD centered around GM. However, these three potential mechanisms are based solely on a comparative analysis of various literature sources, and their validity and feasibility require further verification through in-depth animal experiments. Figure 2 illustrates the brief mechanism of action between GM and IDD.

Figure 2 Possible mechanism of GM-mediated IDD. GM imbalance can accelerate the occurrence and development of IDD through immune regulation, bacterial translocation and colonization, and decomposition and absorption of intestinal metabolites.

Modern Research Progress on Targeting GM to Delay IDD

As the largest avascular tissues in the human body, IVDs rely on a crucial nutrient supply for effective function. Metabolic imbalance, ECM component changes, and IVD cell senescence and apoptosis are significant risk factors for IDD. There are numerous treatment options for IDD, primarily surgical and conservative treatments. Surgical treatment can alleviate symptoms such as compression and pain but cannot fundamentally reverse the biomechanical disorders caused by spinal degeneration. Conservative treatment can alleviate some clinical symptoms of IDD patients, but its effectiveness varies, and the treatment course is relatively long. Both treatments cannot effectively reverse the biological progression of IDD.58 The following is the first review of the potential therapeutic modalities targeting GM to delay IDD in recent years, so as to provide clinical reference.

In recent years, GM colonization has attracted extensive attention as a promising therapeutic target. Lactobacillus paracasei (L. paracasei), mainly in the human oral cavity and intestines, alleviates inflammation-related diseases by regulating anti-inflammatory and pro-inflammatory cytokine secretion.59 Research has shown that colonizing L. paracasei S16 in IDD mice promotes IVD cell proliferation, reduces cell apoptosis, and significantly improves the behavioral scores of IDD mice compared to the non-intervention group. Furthermore, it alleviates the abnormal inflammatory response in IDD mice.60 Unlike targeting a single microbial community, fecal microbiota transplantation (FMT) involves colonizing the patient’s gut with a suspension of feces from a healthy individual to alter the composition and abundance of the host’s GM and restore gut microbiota balance.61 Compared to a single microbial community, this approach maximizes the maintenance of the host’s GM and metabolite integrity, thereby more effectively restoring GM dynamic balance. It has been reported that in an IDD rat model, FMT treatment via fecal suspension gavage not only significantly reduces histopathological damage to IVD tissues, decreases the secretion of inflammatory factors such as TNF-α, IL-1β, IL-6, MMP-3, and MMP-13, but also maintains the stability of IVD cells. Additionally, compared to the IDD group, the FMT intervention group showed improved GM abundance and diversity in rats.62 This indicates that FMT could effectively target and slow IDD progression. However, research on GM colonization for treating IDD is still limited, and there is controversy over the duration of the effects of this method in slowing IDD progression. Besides GM colonization to slow IDD, researchers are also targeting intracellular modification processes to influence GM ecological balance. N6-methyladenosine (m6A), the most common internal modification of mammalian mRNA, affects gene expression in various transcriptional processes. Methyltransferase-like 3 (METTL3) is a key protein in the m6A methyltransferase complex with specific catalytic activity that regulates RNA splicing and assists in the regulation of various physiological and pathological processes.63 Studies have found that METTL3 is highly expressed in IDD rats, and knocking out METTL3 can restore specific gut microbiota levels in IDD rats while altering their pathological phenotype. Moreover, inhibiting METTL3 significantly improves IVD cell senescence and damage, potentially involving TLR2 m6A modification and gut microbiota regulation.64 This suggests that targeting GM to inhibit METTL3 could be a potential therapeutic target for delaying IDD progression.

The development of theories such as the gut-bone axis and gut-disc axis in modern medicine aligns with the holistic concepts of traditional Chinese medicine (TCM). For millennia, Chinese herbal medicines have been employed in Asian countries such as China, Japan, and Korea to treat various orthopedic diseases, including IDD. Recently, researchers have targeted GM with Chinese herbal medicines to inhibit the inflammatory cascade within IVDs, effectively slowing the progression of IDD. Sanbi Decoction, which was first documented in the Song Dynasty’s “Women’s Comprehensive Prescriptions”, has been primarily used for the treatment of degenerative spinal diseases for a long time. Research found that it can broadly regulate GM and serum metabolic homeostasis, reduce inflammatory responses, inhibit ECM degradation, restore IVD height and water content, and significantly slow IDD progression.65 Similarly, the Qiangjin Zhuanggu Qufeng Mixture can maintain the balance between ECM synthesis and degradation, thus slowing the progression of IDD. This process may involve improving the abundance of GM bacteria like Enterobacteriaceae and Clostridium butyricum, inhibiting NLRP3 inflammasome expression, and preventing nucleus pulposus cell pyroptosis.66 Additionally, other studies have shown that Yaobishu Capsules and their active components, palmitic acid (PA) and trans-4-methoxycinnamic acid (THMC), can reshape GM balance after IDD in rats, restore metabolic homeostasis, activate autophagy and the Wnt/β-catenin pathway, reduce IDD-induced inflammation and pain, thereby slowing the progression of IDD.67 Table 1 systematically summarizes three potential therapeutic approaches targeting GM to slow IDD progression.

Table 1 Potential Therapeutic Approaches Targeting GM to Delay IDD

Discussion

Intestinal dysbiosis is a pathological state characterized by the excessive growth of pathogenic microorganisms in the host. Research has shown that this process is linked to the onset and progression of various inflammatory and immune diseases. However, the potential mechanisms of interaction between GM and IDD remain unclear. Taking the gut - disc axis as the starting point, this paper first summarizes the connections between GM imbalance and human diseases, then comprehensively reviews GM’s regulatory mechanisms in skeletal diseases, and finally systematically summarizes the current potential therapies targeting the gut - disc axis to delay IDD for the first time. We found that GM imbalance and disordered metabolite accumulation affect the host’s intestinal balance and mediate systemic diseases, including nervous, cardiovascular, and endocrine systems. Similar findings demonstrated that gut microbiota dysbiosis correlates with increased inflammatory markers associated with systemic diseases.68,69 GM dysbiosis can also accelerate IDD development through immune regulation, bacterial translocation and colonization, and intestinal metabolite breakdown and absorption. In line with this, Studies have highlighted that the inflammatory response elicited by gut dysbiosis contributes significantly to the pathophysiology of disc degeneration.70 Moreover, the current therapeutic strategies targeting the gut-disc axis to delay IDD can be categorized into three types: gut microbiota colonization, intracellular modification regulation, and traditional Chinese medicine intervention. It is important to note that even the drugs referenced for addressing IDD in this manuscript are based on preclinical studies, which may not result in clinically significant or favorable outcomes. Based on these findings, we believe targeting GM and IVD microbiome inflammation and interrupting the cascade reaction in IVDs may effectively delay IDD progression. Therefore, comprehensive and systematic research on the impact of GM on IDD will significantly broaden our understanding of intestinal microbial ecology and skeletal diseases, offering new perspectives for the prevention and treatment of skeletal diseases under precision medicine.

However, research in this area still has limitations: Currently, most of the literature cited in this article relates to preclinical models, which may not indicate a direct causal relationship between the GM and IDD. Additionally, studies have shown that interventions effective in animal models do not always translate to human subjects, suggesting a need for careful consideration of species differences in future research. As the study highlights, the human microbiome is diverse, stable and resilient, and the differences between animal models complicate the extrapolation of results.71 Moreover, due to factors such as cost, time, and participation, studies on the relationship between GM and IDD are scarce and primarily limited to animal experiments. Consequently, clinical applications remain unlikely at present. Additionally, the interactions between the gut ecosystem and the host are often multifactorial, with unknown confounding variables potentially influencing the conclusions drawn from these studies. Thus, further investigations are necessary to explore the benefits of gut microbiome interventions and to elucidate the detailed mechanisms by which GM influences spinal disease progression.

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

This article was supported by National Natural Science Foundation of China (82174402).

Disclosure

The authors declare that they have no competing interests in this work.

References

1. Wang Z, Li X, Yu P, et al. Role of autophagy and pyroptosis in intervertebral disc degeneration. J Inflamm Res. 2024;17:91–100. doi:10.2147/JIR.S434896

2. Sudo H. Intervertebral disc degeneration and regeneration: new molecular mechanisms and therapeutics. Cells. 2024;13(2):153. doi:10.3390/cells13020153

3. de Vos WM, Tilg H, Van Hul M, Cani PD. Gut microbiome and health: mechanistic insights. Gut. 2022;71(5):1020–1032. doi:10.1136/gutjnl-2021-326789

4. Zheng L, Shen J, Chen R, et al. Genome engineering of the human gut microbiome. J Genet Genomics. 2024;51(5):479–491. doi:10.1016/j.jgg.2024.01.002

5. Chen Y, Zhou J, Wang L. Role and mechanism of gut microbiota in human disease. Front Cell Infect Microbiol. 2021;11:625913. doi:10.3389/fcimb.2021.625913

6. Pickard JM, Zeng MY, Caruso R, Núñez G. Gut microbiota: role in pathogen colonization, immune responses, and inflammatory disease. Immunol Rev. 2017;279(1):70–89. doi:10.1111/imr.12567

7. Martin-Gallausiaux C, Marinelli L, Blottière HM, Larraufie P, Lapaque N. SCFA: mechanisms and functional importance in the gut. Proc Nutr Soc. 2021;80(1):37–49. doi:10.1017/S0029665120006916

8. Li J, Zhao F, Wang Y, et al. Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome. 2017;5(1):14. doi:10.1186/s40168-016-0222-x

9. Yan Q, Gu Y, Li X, et al. Alterations of the gut microbiome in hypertension. Front Cell Infect Microbiol. 2017;7:381. doi:10.3389/fcimb.2017.00381

10. Campbell C, Kandalgaonkar MR, Golonka RM, Yeoh BS, Vijay-Kumar M, Saha P. Crosstalk between gut microbiota and host immunity: impact on inflammation and immunotherapy. Biomedicines. 2023;11(2):294. doi:10.3390/biomedicines11020294

11. Liu H, Chen X, Hu X, et al. Alterations in the gut microbiome and metabolism with coronary artery disease severity. Microbiome. 2019;7(1):68. doi:10.1186/s40168-019-0683-9

12. Heintz-Buschart A, Pandey U, Wicke T, et al. The nasal and gut microbiome in Parkinson’s disease and idiopathic rapid eye movement sleep behavior disorder. Mov Disord. 2018;33(1):88–98. doi:10.1002/mds.27105

13. Qian Y, Yang X, Xu S, et al. Alteration of the fecal microbiota in Chinese patients with Parkinson’s disease. Brain Behav Immun. 2018;70:194–202. doi:10.1016/j.bbi.2018.02.016

14. Angelucci F, Cechova K, Amlerova J, Hort J. Antibiotics, gut microbiota, and Alzheimer’s disease. J Neuroinflammation. 2019;16(1):108. doi:10.1186/s12974-019-1494-4

15. Liu S, Gao J, Zhu M, Liu K, Zhang HL. Gut microbiota and dysbiosis in Alzheimer’s disease: implications for pathogenesis and treatment. Mol Neurobiol. 2020;57(12):5026–5043. doi:10.1007/s12035-020-02073-3

16. Zhuang ZQ, Shen LL, Li WW, et al. Gut microbiota is altered in patients with Alzheimer’s disease. J Alzheimers Dis. 2018;63(4):1337–1346. doi:10.3233/JAD-180176

17. Qi X, Yun C, Pang Y, Qiao J. The impact of the gut microbiota on the reproductive and metabolic endocrine system. Gut Microbes. 2021;13(1):1–21. doi:10.1080/19490976.2021.1894070

18. Gomes AC, Hoffmann C, Mota JF. The human gut microbiota: metabolism and perspective in obesity. Gut Microbes. 2018;9(4):308–325. doi:10.1080/19490976.2018.1465157

19. Zhao L, Lou H, Peng Y, et al. Comprehensive relationships between gut microbiome and faecal metabolome in individuals with type 2 diabetes and its complications. Endocrine. 2019;66(3):526–537. doi:10.1007/s12020-019-02103-8

20. Gou W, Ling CW, He Y, et al. Interpretable machine learning framework reveals robust gut microbiome features associated with type 2 diabetes. Diabetes Care. 2021;44(2):358–366. doi:10.2337/dc20-1536

21. Lu L, Chen X, Liu Y, Yu X. Gut microbiota and bone metabolism. FASEB J. 2021;35(7). doi:10.1096/fj.202100451R

22. Inchingolo F, Inchingolo AM, Piras F, et al. The interaction between gut microbiome and bone health. Curr Opin Endocrinol Diabetes Obes. 2024;31(3):122–130. doi:10.1097/MED.0000000000000863

23. Rajasekaran S, Soundararajan DCR, Tangavel C, et al. Human intervertebral discs harbour a unique microbiome and dysbiosis determines health and disease. Eur Spine J. 2020;29(7):1621–1640. doi:10.1007/s00586-020-06446-z

24. Li W, Lai K, Chopra N, Zheng Z, Das A, Diwan AD. Gut-disc axis: a cause of intervertebral disc degeneration and low back pain? Eur Spine J. 2022;31(4):917–925. doi:10.1007/s00586-022-07152-8

25. Guo YF, Su T, Yang M, et al. The role of autophagy in bone homeostasis. J Cell Physiol. 2021;236(6):4152–4173. doi:10.1002/jcp.30111

26. Choi JY. Healthy bone tissue homeostasis. Exp Mol Med. 2020;52(8):1165. doi:10.1038/s12276-020-0472-3

27. Kim JM, Lin C, Stavre Z, Greenblatt MB, Shim JH. Osteoblast-osteoclast communication and bone homeostasis. Cells. 2020;9(9):2073. doi:10.3390/cells9092073

28. Tang Y, Peng B, Liu J, et al. Systemic immune-inflammation index and bone mineral density in postmenopausal women: a cross-sectional study of the national health and nutrition examination survey (NHANES) 2007–2018. Front Immunol. 2022;13:975400. doi:10.3389/fimmu.2022.975400

29. Wu D, Cline-Smith A, Shashkova E, et al. T-cell mediated inflammation in postmenopausal osteoporosis. Front Immunol. 2021;12:687551. doi:10.3389/fimmu.2021.687551

30. Yan J, Takakura A, Zandi-Nejad K, Charles JF. Mechanisms of gut microbiota-mediated bone remodeling. Gut Microbes. 2018;9(1):84–92. doi:10.1080/19490976.2017.1371893

31. Wallimann A, Magrath W, Thompson K, et al. Gut microbial-derived short-chain fatty acids and bone: a potential role in fracture healing. Eur Cells Mater. 2021;41:454–470. doi:10.22203/eCM.v041a29

32. Lorenzo J. From the gut to bone: connecting the gut microbiota with Th17 T lymphocytes and postmenopausal osteoporosis. J Clin Invest. 2021;131(5). doi:10.1172/JCI146619

33. Hartmann B, Longo M, Mathiesen DS, et al. Signs of a glucose- and insulin-independent gut-bone axis and aberrant bone homeostasis in type 1 diabetes. J Clin Endocrinol Metab. 2023;109(1):e259–e265. doi:10.1210/clinem/dgad431

34. Nauck MA, Meier JJ. The incretin effect in healthy individuals and those with type 2 diabetes: physiology, pathophysiology, and response to therapeutic interventions. Lancet Diabetes Endocrinol. 2016;4(6):525–536. doi:10.1016/S2213-8587(15)00482-9

35. Fournier DE, Kiser PK, Shoemaker JK, Battié MC, Séguin CA. Vascularization of the human intervertebral disc: a scoping review. JOR Spine. 2020;3(4). doi:10.1002/jsp2.1123

36. Cosamalón-Gan I, Cosamalón-Gan T, Mattos-Piaggio G, et al. Inflammation in the intervertebral disc herniation. Inflamación en la hernia del disco intervertebral. Neurocirugia. 2021;32(1):21–35. doi:10.1016/j.neucir.2020.01.001

37. Ravichandran D, Pillai J, Krishnamurthy K. Genetics of intervertebral disc disease: a review. Clin Anat. 2022;35(1):116–120. doi:10.1002/ca.23803

38. Song C, Zhou Y, Cheng K, et al. Cellular senescence - molecular mechanisms of intervertebral disc degeneration from an immune perspective. Biomed Pharmacother. 2023;162:114711. doi:10.1016/j.biopha.2023.114711

39. Ye F, Lyu FJ, Wang H, Zheng Z. The involvement of immune system in intervertebral disc herniation and degeneration. JOR Spine. 2022;5(1). doi:10.1002/jsp2.1196

40. Chen X, Wang Z, Deng R, et al. Intervertebral disc degeneration and inflammatory microenvironment: expression, pathology, and therapeutic strategies. Inflamm Res. 2023;72(9):1811–1828. doi:10.1007/s00011-023-01784-2

41. Li Z, Yang H, Hai Y, Cheng Y. Regulatory effect of inflammatory mediators in intervertebral disc degeneration. Mediators Inflamm. 2023;2023:6210885. doi:10.1155/2023/6210885

42. 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

43. Mazmanian SK, Liu CH, Tzianabos AO, Kasper DL. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell. 2005;122(1):107–118. doi:10.1016/j.cell.2005.05.007

44. Weiss GA, Hennet T. Mechanisms and consequences of intestinal dysbiosis. Cell Mol Life Sci. 2017;74(16):2959–2977. doi:10.1007/s00018-017-2509-x

45. Yoo JY, Groer M, Dutra SV, Sarkar A, McSkimming DI. Gut microbiota and immune system interactions. Microorganisms. 2020;8(10):1587. doi:10.3390/microorganisms8101587

46. Risbud MV, Shapiro IM. Role of cytokines in intervertebral disc degeneration: pain and disc content. Nat Rev Rheumatol. 2014;10(1):44–56. doi:10.1038/nrrheum.2013.160

47. Huygen FJPM, Kallewaard JW, Nijhuis H, et al. Effectiveness and safety of dorsal root ganglion stimulation for the treatment of chronic pain: a pooled analysis. Neuromodulation. 2020;23(2):213–221. doi:10.1111/ner.13074

48. Dourado E, Ferro M, Sousa Guerreiro C, Fonseca JE. Diet as a modulator of intestinal microbiota in Rheumatoid arthritis. Nutrients. 2020;12(11):3504. doi:10.3390/nu12113504

49. Waldbaum JDH, Xhumari J, Akinsuyi OS, et al. Association between dysbiosis in the gut microbiota of primary osteoporosis patients and bone loss. Aging Dis. 2023;14(6):2081–2095. doi:10.14336/AD.2023.0425

50. Thevaranjan N, Puchta A, Schulz C, et al. Age-associated microbial dysbiosis promotes intestinal permeability, systemic inflammation, and macrophage dysfunction. Cell Host Microbe. 2017;21(4):455–466.e4. doi:10.1016/j.chom.2017.03.002

51. Schirmer M, Smeekens SP, Vlamakis H, et al. Linking the human gut microbiome to inflammatory cytokine production capacity. Cell. 2016;167(4):1125–1136.e8. doi:10.1016/j.cell.2016.10.020

52. Umale S, Yoganandan N. Mechanisms of cervical spine disc injury under cyclic loading. Asian Spine J. 2018;12(5):910–918. doi:10.31616/asj.2018.12.5.910

53. Mann ER, Lam YK, Uhlig HH. Short-chain fatty acids: linking diet, the microbiome and immunity. Nat Rev Immunol. 2024;24:577–595. doi:10.1038/s41577-024-01014-8

54. Portincasa P, Bonfrate L, Vacca M, et al. Gut microbiota and short chain fatty acids: implications in glucose homeostasis. Int J Mol Sci. 2022;23(3):1105. doi:10.3390/ijms23031105

55. Ratajczak W, Rył A, Mizerski A, et al. Immunomodulatory potential of gut microbiome-derived short-chain fatty acids (SCFAs). Acta Biochim Pol. 2019;66(1):1–12. doi:10.18388/abp.2018_2648

56. Qiu X, Gui Y, Xu Y, Li D, Wang L. DHEA promotes osteoblast differentiation by regulating the expression of osteoblast-related genes and Foxp3+ regulatory T cells. Biosci Trends. 2015;9(5):307–314. doi:10.5582/bst.2015.01073

57. Grant MP, Epure LM, Bokhari R, Roughley P, Antoniou J, Mwale F. Human cartilaginous endplate degeneration is induced by calcium and the extracellular calcium-sensing receptor in the intervertebral disc. Eur Cells Mater. 2016;32:137–151. doi:10.22203/ecm.v032a09

58. Wu PH, Kim HS, Jang IT. Intervertebral disc diseases part 2: a review of the current diagnostic and treatment strategies for intervertebral disc disease. Int J Mol Sci. 2020;21(6):2135. doi:10.3390/ijms21062135

59. Cao J, Liu Q, Hao H, et al. Lactobacillus paracasei X11 ameliorates hyperuricemia and modulates gut microbiota in mice. Front Immunol. 2022;13:940228. doi:10.3389/fimmu.2022.940228

60. Wang Z, Wu H, Chen Y, et al. Lactobacillus paracasei S16 alleviates lumbar disc herniation by modulating inflammation response and gut microbiota. Front Nutr. 2021;8:701644. doi:10.3389/fnut.2021.701644

61. Biazzo M, Deidda G. Fecal microbiota transplantation as new therapeutic avenue for human diseases. J Clin Med. 2022;11(14):4119. doi:10.3390/jcm11144119

62. Yao B, Cai Y, Wang W, et al. The effect of gut microbiota on the progression of intervertebral disc degeneration. Orthop Surg. 2023;15(3):858–867. doi:10.1111/os.13626

63. Xu W, Li J, He C, et al. METTL3 regulates heterochromatin in mouse embryonic stem cells. Nature. 2021;591(7849):317–321. doi:10.1038/s41586-021-03210-1

64. Ni S, Huang X, Li X, et al. METTL3 promotes nucleus pulposus cell senescence in intervertebral disc degeneration by regulating TLR2 m6A methylation and gut microbiota. J Gerontol a Biol Sci Med Sci. 2024;79:glae150. doi:10.1093/gerona/glae150

65. Wang N, Chen S, Xie Y, et al. The Sanbi decoction alleviates intervertebral disc degeneration in rats through intestinal flora and serum metabolic homeostasis modulation. Phytomedicine. 2024;127:155480. doi:10.1016/j.phymed.2024.155480

66. Wang D, Zhang L, He D, et al. Systemic pharmacology reveal the mechanism by which the Qiangjin Zhuanggu Qufeng mixture inhibits LPS-induced pyroptosis of rat nucleus pulposus cells. Phytomedicine. 2023;119:154998. doi:10.1016/j.phymed.2023.154998

67. Li S, Chen T, Zhou Y, Li X. Palmitic acid and trans-4-hydroxy-3-methoxycinnamate, the active ingredients of Yaobishu formula, reduce inflammation and pain by regulating gut microbiota and metabolic changes after lumbar disc herniation to activate autophagy and the Wnt/β-catenin pathway. Biochim Biophys Acta Mol Basis Dis. 2024;1870(2):166972. doi:10.1016/j.bbadis.2023.166972

68. Mou Y, Du Y, Zhou L, et al. Gut microbiota interact with the brain through systemic chronic inflammation: implications on neuroinflammation, neurodegeneration, and aging. Front Immunol. 2022;13:796288. doi:10.3389/fimmu.2022.796288

69. Kataoka K. The intestinal microbiota and its role in human health and disease. J Med Invest. 2016;63(1–2):27–37. doi:10.2152/jmi.63.27

70. Rajasekaran S, Vasudevan G, Tangavel C, et al. Does the gut microbiome influence disc health and disease? The interplay between dysbiosis, pathobionts, and disc inflammation: a pilot study. Spine J. 2024;24(10):1952–1963. doi:10.1016/j.spinee.2024.06.020

71. Lozupone CA, Stombaugh JI, Gordon JI, Jansson JK, Knight R. Diversity, stability and resilience of the human gut microbiota. Nature. 2012;489(7415):220–230. doi:10.1038/nature11550

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.