Acessibilidade / Reportar erro

Impact of gut-peripheral nervous system axis on the development of diabetic neuropathy

Abstract

Diabetes is a chronic metabolic disease caused by a reduction in the production and/or action of insulin, with consequent development of hyperglycemia. Diabetic patients, especially those who develop neuropathy, presented dysbiosis, with an increase in the proportion of pathogenic bacteria and a decrease in the butyrate-producing bacteria. Due to this dysbiosis, diabetic patients presented a weakness of the intestinal permeability barrier and high bacterial product translocation to the bloodstream, in parallel to a high circulating levels of pro-inflammatory cytokines such as TNF-α. In this context, we propose here that dysbiosis-induced increased systemic levels of bacterial products, like lipopolysaccharide (LPS), leads to an increase in the production of pro-inflammatory cytokines, including TNF-α, by Schwann cells and spinal cord of diabetics, being crucial for the development of neuropathy.

Key words:
diabetes; dysbiosis; gut microbiota; inflammation; neuropathy


Diabetes is a chronic metabolic disease characterized by hyperglycemia due to a reduction in the production and/or action of insulin.11. Insuela DBR, Ferrero MR, Gonçalves-de-Albuquerque CF, Chaves ADS, da Silva AYO, Castro-Faria-Neto HC, et al. Glucagon reduces neutrophil migration and increases susceptibility to sepsis in diabetic mice. Front Immunol. 2021; 12: 633540. Currently, diabetes is one of the most serious and frequent chronic diseases in worldwide. Uncontrolled diabetes is accompanied by the development of several disabling and costly complications, which reduce patients’ life expectancy and can be fatal.22. Heald AH, Stedman M, Davies M, Livingston M, Alshames R, Lunt M, et al. Estimating life years lost to diabetes: outcomes from analysis of National Diabetes Audit and Office of National Statistics data. Cardiovasc Endocrinol Metab. 2020; 9(4): 183-5. In 2021, the global prevalence of diabetes reached pandemic proportions with 537 million people living with diabetes in the world, accompanied by an expense of 699 billion USD in global healthcare. In addition, future projections suggest that up to 2045 the number of people with diabetes will increase by 46%, with an estimate of health expenditures for the care of this disease that will exceed one trillion USD.33. Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al. IDF diabetes atlas: global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022; 183: 109119.

Neuropathy is the most prevalent complication of diabetes, occurring in up to half of all people living with this disease.44. Feldman EL, Callaghan BC, Pop-Busui R, Zochodne DW, Wright DE, Bennett DL, et al. Diabetic neuropathy. Nat Rev Dis Primers. 2019; 5(1): 41. In addition, neuropathy is responsible for frequent hospitalization compared to other diabetes morbidities,55. Patel S, Farkash C, Simmons D. Type 1 diabetes management and hospitalisation in the over 25's at an Australian outer urban diabetes clinic. BMC Endocr Disord. 2022; 22(1): 143. and it is the most common reason for non-traumatic amputation.66. Hicks CW, Selvin E. Epidemiology of peripheral neuropathy and lower extremity disease in diabetes. Curr Diab Rep. 2019; 19(10): 86. Neuropathic pain is manifested as spontaneous or induced pain, such as hyperalgesia and allodynia.77. Kamerman PR, Wadley AL, Davis KD, Hietaharju A, Jain P, Kopf A, et al. World Health Organization essential medicines lists: where are the drugs to treat neuropathic pain? Pain. 2015; 156(5): 793-7.,88. Finnerup NB, Attal N, Haroutounian S, McNicol E, Baron R, Dworkin RH, et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol. 2015; 14(2): 162-73. Although neuropathy is the strongest predictor of mortality in diabetes, it remains without specific treatment.44. Feldman EL, Callaghan BC, Pop-Busui R, Zochodne DW, Wright DE, Bennett DL, et al. Diabetic neuropathy. Nat Rev Dis Primers. 2019; 5(1): 41. This scenario leads to high individual costs for patients, including pain, inability to work, poor quality of life, multiple hospitalizations for ulcers and eventual amputations. Therefore, we performed a narrative review with the aim of increasing knowledge about the role of gut dysbiosis in the development and/or progression of neuropathy in diabetic patients, besides how essential this microenvironment is for better control of the disease.

Pathogenesis of diabetic neuropathy

In diabetic patients, the development of neuropathy is multifactorial and involves uncontrolled glycemia, diabetes duration, and age-related neuronal attrition.44. Feldman EL, Callaghan BC, Pop-Busui R, Zochodne DW, Wright DE, Bennett DL, et al. Diabetic neuropathy. Nat Rev Dis Primers. 2019; 5(1): 41.,66. Hicks CW, Selvin E. Epidemiology of peripheral neuropathy and lower extremity disease in diabetes. Curr Diab Rep. 2019; 19(10): 86. Although the precise order of cellular injury in diabetes is unknown, the alterations in the peripheral nervous system that culminate in diabetic neuropathy are well established. These changes include (i) progressive loss of neurofilament polymer, which are essential structural scaffolds of the axon;99. Scott JN, Clark AW, Zochodne DW. Neurofilament and tubulin gene expression in progressive experimental diabetes: failure of synthesis and export by sensory neurons. Brain. 1999; 122(11): 2109-18. (ii) modification in the key plasticity molecules in the dorsal root ganglia (DRG), including decrease in the synthesis of growth-associated protein 43 (GAP43) and β-tubulin and increase in the expression of heat shock proteins (HSP) and poly(ADP-ribose) polymerase (PARP);1010. Ma J, Pan P, Anyika M, Blagg BS, Dobrowsky RT. Modulating molecular chaperones improves mitochondrial bioenergetics and decreases the inflammatory transcriptome in diabetic sensory neurons. ACS Chem Neurosci. 2015; 6(9): 1637-48.,1111. Urban MJ, Pan P, Farmer KL, Zhao H, Blagg BS, Dobrowsky RT. Modulating molecular chaperones improves sensory fiber recovery and mitochondrial function in diabetic peripheral neuropathy. Exp Neurol. 2012; 235(1): 388-96.,1212. Ilnytska O, Lyzogubov VV, Stevens MJ, Drel VR, Mashtalir N, Pacher P, et al. Poly(ADP-ribose) polymerase inhibition alleviates experimental diabetic sensory neuropathy. Diabetes. 2006; 55(6): 1686-94.,1313. Lupachyk S, Shevalye H, Maksimchyk Y, Drel VR, Obrosova IG. PARP inhibition alleviates diabetes-induced systemic oxidative stress and neural tissue 4-hydroxynonenal adduct accumulation: correlation with peripheral nerve function. Free Radic Biol Med. 2011; 50(10): 1400-9. (iii) axonal degeneration;1414. Cashman CR, Höke A. Mechanisms of distal axonal degeneration in peripheral neuropathies. Neurosci Lett. 2015; 596: 33-50. (iv) reduction in the Schwann cells to support axons, through decrease in the provision of cytoskeletal support, trophic factors or ribosome transfer that allows intra-axonal mRNA translation within distal axons;1515. Feldman EL, Nave KA, Jensen TS, Bennett DLH. New horizons in diabetic neuropathy: mechanisms, bioenergetics, and pain. Neuron. 2017; 93(6): 1296-1313. (v) demyelination, which occurs in more severe cases of diabetic neuropathy;1616. Dunnigan SK, Ebadi H, Breiner A, Katzberg HD, Lovblom LE, Perkins BA, et al. Conduction slowing in diabetic sensorimotor polyneuropathy. Diabetes Care. 2013; 36(11): 3684-90.,1717. Gumy LF, Bampton ET, Tolkovsky AM. Hyperglycaemia inhibits Schwann cell proliferation and migration and restricts regeneration of axons and Schwann cells from adult murine DRG. Mol Cell Neurosci. 2008; 37(2): 298-311.,1818. Mizisin AP, Shelton GD, Wagner S, Rusbridge C, Powell HC. Myelin splitting, Schwann cell injury and demyelination in feline diabetic neuropathy. Acta Neuropathol. 1998; 95(2): 171-4. (vi) reduction in the blood flow in the DRG1919. Zochodne DW, Ho LT. The influence of sulindac on experimental streptozotocin-induced diabetic neuropathy. Can J Neurol Sci. 1994; 21(3): 194-202. (Fig. 1).

Fig. 1:
alterations observed in the peripheral nervous system in diabetic neuropathy. In the dorsal root ganglion (DRG), localization of the cell bodies of sensory neurons, diabetes induced alterations in the key plasticity molecules, including reduction in growth-associated protein 43 (GAP43) and β-tubulin in parallel with an increase in the heat shock proteins (HSP) and poly(ADP-ribose) polymerase (PARP) expression. Furthermore, diabetics showed loss of neurofilament polymer, decrease in the Schwann cells-axon support, axonal degeneration, and ischemia in the sensory nerves, in addition to a demyelination in more severe cases of diabetic neuropathy. Altogether, these alterations evoked pain in diabetic patients.

The exact mechanism that promotes alterations in the peripheral nervous system and, consequently, neuropathic pain in diabetic patients is not fully elucidated. Nonetheless, several evidences showed that hyperglycemia and dyslipidemia induce pathological changes in neurons, glia, and vascular cells that culminate in nerve dysfunction and neuropathy.44. Feldman EL, Callaghan BC, Pop-Busui R, Zochodne DW, Wright DE, Bennett DL, et al. Diabetic neuropathy. Nat Rev Dis Primers. 2019; 5(1): 41. Hyperglycemia increases glycolysis, polyol, advanced glycation end products, protein kinase C, and hexosamine pathways in Schwann cells, DRG neurons and axons.1616. Dunnigan SK, Ebadi H, Breiner A, Katzberg HD, Lovblom LE, Perkins BA, et al. Conduction slowing in diabetic sensorimotor polyneuropathy. Diabetes Care. 2013; 36(11): 3684-90.,1717. Gumy LF, Bampton ET, Tolkovsky AM. Hyperglycaemia inhibits Schwann cell proliferation and migration and restricts regeneration of axons and Schwann cells from adult murine DRG. Mol Cell Neurosci. 2008; 37(2): 298-311.,1818. Mizisin AP, Shelton GD, Wagner S, Rusbridge C, Powell HC. Myelin splitting, Schwann cell injury and demyelination in feline diabetic neuropathy. Acta Neuropathol. 1998; 95(2): 171-4.,2020. Kang Q, Yang C. Oxidative stress and diabetic retinopathy: molecular mechanisms, pathogenetic role and therapeutic implications. Redox Biol. 2020; 37: 101799. These metabolic changes result in increased reactive oxygen species (ROS) formation and release of pro-inflammatory mediators. In parallel, the dyslipidemia observed in diabetic patients also induced a rise in ROS production and systemic and local inflammation.2121. Jang ER, Lee CS. 7-ketocholesterol induces apoptosis in differentiated PC12 cells via reactive oxygen species-dependent activation of NF-?B and Akt pathways. Neurochem Int. 2011; 58(1): 52-9.,2222. Legrand-Poels S, Esser N, L'homme L, Scheen A, Paquot N, Piette J. Free fatty acids as modulators of the NLRP3 inflammasome in obesity/type 2 diabetes. Biochem Pharmacol. 2014; 92(1): 131-41.

In diabetes, the overproduction of ROS induces activation of PARP-1 with a concomitant decrease in ATP formation.2323. Burkart V, Wang ZQ, Radons J, Heller B, Herceg Z, Stingl L, et al. Mice lacking the poly(ADP-ribose) polymerase gene are resistant to pancreatic beta-cell destruction and diabetes development induced by streptozocin. Nat Med. 1999; 5(3): 314-9. Altogether, high levels of ROS and loss of ATP production cause mitochondrial failure and metabolic and oxidative damage in Schwann cells and DRG neurons.2424. Fernyhough P, McGavock J. Mechanisms of disease: mitochondrial dysfunction in sensory neuropathy and other complications in diabetes. Handb Clin Neurol. 2014; 126: 353-77.,2525. Chowdhury SK, Smith DR, Fernyhough P. The role of aberrant mitochondrial bioenergetics in diabetic neuropathy. Neurobiol Dis. 2013; 51: 56-65. The increase in ROS formation, dysfunctional mitochondria, and microvascular insufficiency result in axonal degeneration in the peripheral nervous system.2626. Huvers FC, De Leeuw PW, Houben AJ, De Haan CH, Hamulyak K, Schouten H, et al. Endothelium-dependent vasodilatation, plasma markers of endothelial function, and adrenergic vasoconstrictor responses in type 1 diabetes under near-normoglycemic conditions. Diabetes. 1999; 48(6): 1300-7.,2727. Rumora AE, Lentz SI, Hinder LM, Jackson SW, Valesano A, Levinson GE, et al. Dyslipidemia impairs mitochondrial trafficking and function in sensory neurons. FASEB J. 2018; 32(1): 195-207.,2828. Hink U, Li H, Mollnau H, Oelze M, Matheis E, Hartmann M, et al. Mechanisms underlying endothelial dysfunction in diabetes mellitus. Circ Res. 2001; 88(2): e14-e22.,2929. Vinik A, Ullal J, Parson HK, Casellini CM. Diabetic neuropathies: clinical manifestations and current treatment options. Nat Clin Pract Endocrinol Metab. 2006; 2(5): 269-81. In addition, the oxidative damage induces hyperexcitability in the axons and DRG neurons, causing neuropathic pain.3030. Kasznicki J, Kosmalski M, Sliwinska A, Mrowicka M, Stanczyk M, Majsterek I, et al. Evaluation of oxidative stress markers in pathogenesis of diabetic neuropathy. Mol Biol Rep. 2012; 39(9): 8669-78.

Several pre-clinical and clinical studies showed upregulation of pathways involved in inflammation in peripheral nerves.3131. Hur J, O'Brien PD, Nair V, Hinder LM, McGregor BA, Jagadish HV, et al. Transcriptional networks of murine diabetic peripheral neuropathy and nephropathy: common and distinct gene expression patterns. Diabetologia. 2016; 59(6): 1297-306.,3232. McGregor BA, Eid S, Rumora AE, Murdock B, Guo K, de Anda-Jáuregui G, et al. Conserved transcriptional signatures in human and murine diabetic peripheral neuropathy. Sci Rep. 2018; 8(1): 17678.,3333. Hur J, Sullivan KA, Pande M, Hong Y, Sima AA, Jagadish HV, et al. The identification of gene expression profiles associated with progression of human diabetic neuropathy. Brain. 2011; 134(Pt 11): 3222-35.,3434. Hur J, Sullivan KA, Callaghan BC, Pop-Busui R, Feldman EL. Identification of factors associated with sural nerve regeneration and degeneration in diabetic neuropathy. Diabetes Care. 2013; 36(12): 4043-9. Furthermore, numerous experimental models of diabetic neuropathy showed an inflammatory response, characterized by infiltration of macrophages and T cells and increased levels of pro-inflammatory cytokines, in the sciatic nerve and DRG.3535. Baum P, Kosacka J, Estrela-Lopis I, Woidt K, Serke H, Paeschke S, et al. The role of nerve inflammation and exogenous iron load in experimental peripheral diabetic neuropathy (PDN). Metabolism. 2016; 65(4): 391-405.,3636. Kosacka J, Woidt K, Toyka KV, Paeschke S, Klöting N, Bechmann I, et al. The role of dietary non-heme iron load and peripheral nerve inflammation in the development of peripheral neuropathy (PN) in obese non-diabetic leptin-deficient ob/ob mice. Neurol Res. 2019; 41(4): 341-53.,3737. Paeschke S, Baum P, Toyka KV, Blüher M, Koj S, Klöting N, et al. The role of iron and nerve inflammation in diabetes mellitus type 2-induced peripheral neuropathy. Neuroscience. 2019; 406: 496-509.,3838. Cheng HT, Dauch JR, Oh SS, Hayes JM, Hong Y, Feldman EL. p38 mediates mechanical allodynia in a mouse model of type 2 diabetes. Mol Pain. 2010; 6: 28. The inflammatory response was accompanied by loss of myelinated and unmyelinated nerve fibers and axonal damage in diabetic animals with neuropathy.3939. Kosacka J, Nowicki M, Klöting N, Kern M, Stumvoll M, Bechmann I, et al. COMP-angiopoietin-1 recovers molecular biomarkers of neuropathy and improves vascularisation in sciatic nerve of ob/ob mice. PLoS One. 2012; 7(3): e32881. The presence of inflammation biomarkers was also associated with onset and progression of neuropathy in diabetic patients.4040. Herder C, Kannenberg JM, Huth C, Carstensen-Kirberg M, Rathmann W, Koenig W, et al. Proinflammatory cytokines predict the incidence and progression of distal sensorimotor polyneuropathy: KORA F4/FF4 study. Diabetes Care. 2017; 40(4): 569-76.,4141. Hall BE, Macdonald E, Cassidy M, Yun S, Sapio MR, Ray P, et al. Transcriptomic analysis of human sensory neurons in painful diabetic neuropathy reveals inflammation and neuronal loss. Sci Rep. 2022; 12(1): 4729. Moreover, diabetes-induced inflammation altered mitochondrial bioenergetics in DRG neurons4242. Chowdhury SK, Zherebitskaya E, Smith DR, Akude E, Chattopadhyay S, Jolivalt CG, et al. Mitochondrial respiratory chain dysfunction in dorsal root ganglia of streptozotocin-induced diabetic rats and its correction by insulin treatment. Diabetes. 2010; 59(4): 1082-91.,4343. Fernyhough P, Roy Chowdhury SK, Schmidt RE. Mitochondrial stress and the pathogenesis of diabetic neuropathy. Expert Rev Endocrinol Metab. 2010; 5(1): 39-49. and HSP content in sensory neurons.4444. Ma J, Farmer KL, Pan P, Urban MJ, Zhao H, Blagg BS, et al. Heat shock protein 70 is necessary to improve mitochondrial bioenergetics and reverse diabetic sensory neuropathy following KU-32 therapy. J Pharmacol Exp Ther. 2014; 348(2): 281-92.,4545. Ma J, Pan P, Anyika M, Blagg BS, Dobrowsky RT. Modulating molecular chaperones improves mitochondrial bioenergetics and decreases the inflammatory transcriptome in diabetic sensory neurons. ACS Chem Neurosci. 2015; 6(9): 1637-48. Besides, the systemic low-grade inflammation has been implicated in neuropathic pain observed in diabetic patients, since some circulating inflammatory mediators was positive related to the severity of neuropathic pain in a subgroup of patients with distal symmetrical polyneuropathy.4646. Bäckryd E, Themistocleous A, Larsson A, Gordh T, Rice AS, Tesfaye S, et al. Hepatocyte growth factor, colony-stimulating factor 1, CD40, and 11 other inflammation-related proteins are associated with pain in diabetic neuropathy: exploration and replication serum data from the Pain in Neuropathy Study. Pain. 2022; 163(5): 897-909.

Diabetes and gut microbiota

The human gut microbiota contains up to 100 trillion of microbes, including commensal, symbiotic, and pathogenic bacteria, as well as archaea, fungi, and viruses.4747. Thursby E, Juge N. Introduction to the human gut microbiota. Biochem J. 2017; 474(11): 1823-36.,4848. Vemuri R, Shankar EM, Chieppa M, Eri R, Kavanagh K. Beyond just bacteria: functional biomes in the gut ecosystem including virome, mycobiome, archaeome and helminths. Microorganisms. 2020; 8(4): 483. Gut microbiota has various physiological functions in the host, including strengthening epithelial-intestinal barrier integrity,4949. Tsang DK, Wang RJ, De Sa O, Ayyaz A, Foerster EG, Bayer G, et al. A single cell survey of the microbial impacts on the mouse small intestinal epithelium. Gut Microbes. 2022; 14(1): 2108281. maintenance of energy homeostasis,5050. Gao Z, Yin J, Zhang J, Ward RE, Martin RJ, Lefevre M, et al. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes. 2009; 58(7): 1509-17. protection against pathogens,5151. Edwinson A, Yang L, Chen J, Grover, M. Colonic expression of Ace2, the SARS-CoV-2 entry receptor, is suppressed by commensal human microbiota. Gut Microbes. 2021; 13(1): 1984105. and regulation of host immunity.5252. Romero R, Zarzycka A, Preussner M, Fischer F, Hain T, Herrmann JP, et al. Selected commensals educate the intestinal vascular and immune system for immunocompetence. Microbiome. 2022; 10(1): 1-18.

Diet, early-life microbiota exposure, antibiotic therapy, changing hygiene status, pollution, socioeconomic status, and other environmental factors can directly influence the composition of gut microbiota and its metabolic products, making it unique to each individua.5353. Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. Human nutrition, the gut microbiome and the immune system. Nature. 2011; 474(7351): 327-36.,5454. Pérez-Cobas AE, Gosalbes MJ, Friedrichs A, Knecht H, Artacho A, Eismann K, et al. Gut microbiota disturbance during antibiotic therapy: a multi-omic approach. Gut. 2013; 62(11): 1591-1601. Furthermore, patients with some pathologic conditions, for instance inflammatory diseases, infections, neurodegenerative diseases, and metabolic diseases, presented an imbalance in the microbes’ composition into the gut with the predominance of pathogenic bacteria, known as dysbiosis.5555. Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017; 17(4): 219-232.,5656. Weiss GA, Hennet T. Mechanisms and consequences of intestinal dysbiosis. Cell Mol Life Sci. 2017; 74(16): 2959-77.

In early stages of life, the extensive exposure to antibiotics may lead to dysbiosis, resulting in underweight or overweight.5757. Weintraub AS, Ferrara L, Deluca L, Moshier E, Green RS, Oakman E, et al. Antenatal antibiotic exposure in preterm infants with necrotizing enterocolitis. J Perinatol. 2012; 32(9): 705-9.,5858. Cotten CM, Taylor S, Stoll B, Goldberg RN, Hansen NI, Sánchez PJ, et al. Prolonged duration of initial empirical antibiotic treatment is associated with increased rates of necrotizing enterocolitis and death for extremely low birth weight infants. Pediatrics. 2009; 123(1): 58-66.,5959. Kim JE, Li B, Fei L, Horne R, Lee D, Loe AK, et al. Gut microbiota promotes stem cell differentiation through macrophage and mesenchymal niches in early postnatal development. Immunity. 2022; 55(12): 2300-17. Moreover, the large consumption of antibiotics is usually been related to the development of metabolic disorders in later stages of life.6060. Li P, Chang X, Chen X, Wang C, Shang Y, Zheng D, et al. Early-life antibiotic exposure increases the risk of childhood overweight and obesity in relation to dysbiosis of gut microbiota: a birth cohort study. Ann Clin Microbiol Antimicrob. 2022; 21(1): 46. In addition, the intake of sweeteners, as sugar substitute, also has considerable impact over microbiota population, inducing glucose intolerance.6161. Ruiz-Ojeda FJ, Plaza-Díaz J, Sáez-Lara MJ, Gil A. Effects of sweeteners on the gut microbiota: a review of experimental studies and clinical trials. Adv Nutr. 2019; 1(10): S31-48.,6262. Li CH, Wang CT, Lin YJ, Kuo HY, Wu JS, Hong TC, et al. Long-term consumption of the sugar substitute sorbitol alters gut microbiome and induces glucose intolerance in mice. Life Sci. 2022; 305: 120770. These observations suggest that an imbalance in the composition of the gut microbiota may be related to the development of metabolic diseases. In fact, the composition of gut microbiota can alter the immune response of non-obese diabetic (NOD) mice, a classical model of autoimmune diabetes. Germ-free (GF) NOD mice showed an acceleration in the insulitis in parallel to a rise in the Th1 and Th17 cells in the mesenteric and pancreatic lymph nodes,6363. Alam C, Bittoun E, Bhagwat D, Valkonen S, Saari A, Jaakkola U, et al. Effects of a germ-free environment on gut immune regulation and diabetes progression in non-obese diabetic (NOD) mice. Diabetologia. 2011; 54(6): 1398-406. however, the incidence of diabetes was not modified.6363. Alam C, Bittoun E, Bhagwat D, Valkonen S, Saari A, Jaakkola U, et al. Effects of a germ-free environment on gut immune regulation and diabetes progression in non-obese diabetic (NOD) mice. Diabetologia. 2011; 54(6): 1398-406.,6464. King C, Sarvetnick N. The incidence of type-1 diabetes in NOD mice is modulated by restricted flora not germ-free conditions. PLoS One. 2011; 6(2): e17049. Furthermore, the administration of probiotics in NOD mice decreased the incidence of diabetes, through reduction of insulitis and gut permeability and modulation of cytokine profile, Treg cells and T helper cell polarization.6565. Calcinaro F, Dionisi S, Marinaro M, Candeloro P, Bonato V, et al. Oral probiotic administration induces interleukin-10 production and prevents spontaneous autoimmune diabetes in the non-obese diabetic mouse. Diabetologia. 2005; 48(8): 1565-75.,6666. Matsuzaki T, Nagata Y, Kado S, Uchida K, Kato I, Hashimoto S, et al. Prevention of onset in an insulin-dependent diabetes mellitus model, NOD mice, by oral feeding of Lactobacillus casei. APMIS. 1997; 105(8): 643-9.,6767. Kim TK, Lee JC, Im SH, Lee MS. Amelioration of autoimmune diabetes of NOD mice by immunomodulating probiotics. Front Immunol. 2020; 11: 1832. These data indicate that gut microbiota is important to the development of diabetes.

It is well known that diabetic patients showed dysbiosis.6868. Durazzo M, Ferro A, Gruden G. Gastrointestinal microbiota and type 1 diabetes mellitus: the state of art. J Clin Med. 2019; 8(11): 1843.,6969. Muñoz-Garach A, Diaz-Perdigones C, Tinahones FJ. Gut microbiota and type 2 diabetes mellitus. Endocrinol Nutr. 2016; 63(10): 560-8. Although the composition of gut microbiota is diverse among diabetic patients, the relationship between Firmicutes and Bacteroidetes is unbalanced in these patients.7070. Santos-Marcos JA, Perez-Jimenez F, Camargo A. The role of diet and intestinal microbiota in the development of metabolic syndrome. J Nutr Biochem. 2019; 70: 1-27.,7171. Ahmad A, Yang W, Chen G, Shafiq M, Javed S, Ali Zaidi SS, et al. Analysis of gut microbiota of obese individuals with type 2 diabetes and healthy individuals. PLoS One. 2019; 14(12): e0226372.,7272. Pascale A, Marchesi N, Govoni S, Coppola A, Gazzaruso C. The role of gut microbiota in obesity, diabetes mellitus, and effect of metformin: new insights into old diseases. Curr Opin Pharmacol. 2019; 49: 1-5. In addition, patients with type 1 diabetes presented a clear depletion of species like Prevotella copri and Bifidobacterium longum, probiotic bacteria, and enrichment of families like Ruminococcaceae, Clostridiaceae, Clostridiales, and Oscillibacter, bacteria associated with infection and inflammation.7373. van Heck JIP, Gacesa R, Stienstra R, Fu J, Zhernakova A, Harmsen HJM, et al. The gut microbiome composition is altered in long-standing type 1 diabetes and associates with glycemic control and disease-related complications. Diabetes Care. 2022; dc212225.

The maintenance of homeostasis in gut environment is important not only to slow-down diabetes development but it is also central in the control of its complications.7474. Snelson M, de Pasquale C, Ekinci EI, Coughlan MT. Gut microbiome, prebiotics, intestinal permeability and diabetes complications. Best Pract Res Clin Endocrinol Metab. 2021; 35(3): 101507. Dysbiosis in diabetic patients and animals lead to an increase in the intestinal permeability7575. Meddings JB, Jarand J, Urbanski SJ, Hardin J, Gall DG. Increased gastrointestinal permeability is an early lesion in the spontaneously diabetic BB rat. Am J Physiol. 1999; 276(4): G951-7.,7676. Carratù R, Secondulfo M, de Magistris L, Iafusco D, Urio A, Carbone MG, et al. Altered intestinal permeability to mannitol in diabetes mellitus type I. J Pediatr Gastroenterol Nutr. 1999; 28(3): 264-9. in parallel to a rise in the bacterial content to the bloodstream, as lipopolysaccharide (LPS).6868. Durazzo M, Ferro A, Gruden G. Gastrointestinal microbiota and type 1 diabetes mellitus: the state of art. J Clin Med. 2019; 8(11): 1843.,7777. Jayashree B, Bibin YS, Prabhu D, Shanthirani CS, Gokulakrishnan K, Lakshmi BS, et al. Increased circulatory levels of lipopolysaccharide (LPS) and zonulin signify novel biomarkers of proinflammation in patients with type 2 diabetes. Mol Cell Biochem. 2014; 388(1-2): 203-10. Interestingly, the supplementation of diabetic mice with a microbial anti-inflammatory molecule, which is a metabolite of a commensal bacteria Faecalibacterium prausnitzii, improved the intestinal barrier permeability and reduced circulating levels of LPS.7878. Xu J, Liang R, Zhang W, Tian K, Li J, Chen X, et al. Faecalibacterium prausnitzii -derived microbial anti-inflammatory molecule regulates intestinal integrity in diabetes mellitus mice via modulating tight junction protein expression. J Diabetes. 2020; 12(3): 224-36.

In diabetic patients, the gut leakiness is accompanied by a low-grade inflammation, with increased levels of IL-1β, IL-6, and TNF-α in the blood.7979. Popko K, Gorska E, Stelmaszczyk-Emmel A, Plywaczewski R, Stoklosa A, Gorecka D, et al. Proinflammatory cytokines Il-6 and TNF-a and the development of inflammation in obese subjects. Eur J Med Res. 2010; 15(Suppl. 2):120-2. Furthermore, type 1 and type 2 diabetic patients as well as NOD mice showed a reduction in the abundance of butyrate-producing bacteria.8080. Ejtahed HS, Hoseini-Tavassol Z, Khatami S, Zangeneh M, Behrouzi A, Ahmadi Badi S, et al. Main gut bacterial composition differs between patients with type 1 and type 2 diabetes and non-diabetic adults. J Diabetes Metab Disord. 2020; 19(1): 265-71.,8181. Ganesan K, Chung SK, Vanamala J, Xu B. Causal relationship between diet-induced gut microbiota changes and diabetes: a novel strategy to transplant Faecalibacterium prausnitzii in preventing diabetes. Int J Mol Sci. 2018; 19(12): 3720. Butyrate is a short-chain fatty acid (SCFA) that induce mucin production, regulating the permeability of the intestinal barrier, and reduce translocation of bacteria and their products, oxidative stress, and inflammation.8282. Guilloteau P, Martin L, Eeckhaut V, Ducatelle R, Zabielski R, Van Immerseel F. From the gut to the peripheral tissues: the multiple effects of butyrate. Nutr Res Rev. 2010; 23(2): 366-84. The reduction of butyrate-forming bacteria, with consequent inadequate butyrate secretion, aggravates the pathogenesis of diabetes,8383. Khan S, Jena G. The role of butyrate, a histone deacetylase inhibitor in diabetes mellitus: experimental evidence for therapeutic intervention. Epigenomics. 2015; 7(4): 669-80. through increase of inflammation and oxidative stress. Treatment with butyrate decreased ROS production and the homeostatic levels of inflammatory markers in diabetic mice.8484. Noureldein MH, Bitar S, Youssef N, Azar S, Eid AA. Butyrate modulates diabetes-linked gut dysbiosis: epigenetic and mechanistic modifications. J Mol Endocrinol. 2020; 64(1): 29-42.

Remarkably, treatment with probiotics is one of the most used strategies to modulate intestinal microbiota and their used can prevent diabetes establishment and is effective as adjuvant in insulin resistance therapies.8585. Andreasen AS, Larsen N, Pedersen-Skovsgaard T, Berg RM, Møller K, Svendsen KD, et al. Effects of Lactobacillus acidophilus NCFM on insulin sensitivity and the systemic inflammatory response in human subjects. Br J Nutr. 2010; 104(12): 1831-8.,8686. Yao K, Zeng L, He Q, Wang W, Lei J, Zou X. Effect of probiotics on glucose and lipid metabolism in type 2 diabetes mellitus: a meta-analysis of 12 randomized controlled trials. Med Sci Monit. 2017; 23: 3044-53.,8787. Alfa MJ, Strang D, Tappia PS, Olson N, DeGagne P, Bray D, et al. A randomized placebo controlled clinical trial to determine the impact of digestion resistant starch MSPrebiotic(r) on glucose, insulin, and insulin resistance in elderly and mid-age adults. Front Med (Lausanne). 2018; 4: 260.,6565. Calcinaro F, Dionisi S, Marinaro M, Candeloro P, Bonato V, et al. Oral probiotic administration induces interleukin-10 production and prevents spontaneous autoimmune diabetes in the non-obese diabetic mouse. Diabetologia. 2005; 48(8): 1565-75.,6666. Matsuzaki T, Nagata Y, Kado S, Uchida K, Kato I, Hashimoto S, et al. Prevention of onset in an insulin-dependent diabetes mellitus model, NOD mice, by oral feeding of Lactobacillus casei. APMIS. 1997; 105(8): 643-9.,6767. Kim TK, Lee JC, Im SH, Lee MS. Amelioration of autoimmune diabetes of NOD mice by immunomodulating probiotics. Front Immunol. 2020; 11: 1832. According to Food and Agriculture Organization (FAO) and World Health Organization (WHO), probiotics are defined as live micro-organisms which when administered in adequate amounts confer a health benefit on the host.8888. FAO/WHO - Food and Agriculture Organization/World Health Organization. Probiotics in food. Health and nutritional properties and guidelines for evaluation. Fao Food and Nutrition. 2006; paper 85. Available from: https://www.fao.org/3/a0512e/a0512e.pdf.
https://www.fao.org/3/a0512e/a0512e.pdf...
Furthermore, treatment of diabetic rats with probiotics slow down the progression of diabetes in clear association with a decrease in the plasma levels of LPS.8989. Wang Y, Liu H, Zheng M, Yang Y, Ren H, Kong Y, et al. Berberine slows the progression of prediabetes to diabetes in zucker diabetic fatty rats by enhancing intestinal secretion of glucagon-like peptide-2 and improving the gut microbiota. Front Endocrinol (Lausanne). 2021; 12: 609134. Besides, the treatment of diabetic animals with probiotics containing the Lactobacillus rhamnosus NCDC17 improved the insulin resistance, in parallel to a reduction in IL-6 and TNF in the epididymal fat.9090. Singh S, Sharma RK, Malhotra S, Pothuraju R, Shandilya UK. Lactobacillus rhamnosus NCDC17 ameliorates type-2 diabetes by improving gut function, oxidative stress and inflammation in high-fat-diet fed and streptozotocintreated rats. Benef Microbes. 2017; 8(2): 243-55. Although the use of adjuvant therapy with probiotics seems to be interesting to treat some comorbidities of diabetes related to dysbiosis, some effects of them are controversy in diabetic patients. In general, the treatment with probiotics is useful to control insulin resistance and improved the intestinal barrier permeability in type 2 diabetic patients, however, it is ineffective in reducing systemic inflammation. Furthermore, the use of probiotics to reduce the glycemia and serum insulin levels in these patients is very controversy. Although the studies with type 1 diabetic patients are still scarce, it was observed that treatment with probiotics reduced glycemia and systemic inflammatory markers as compared to placebo (Table).

TABLE
Clinical trials in diabetic patients after probiotic administration

Gut microbiota and neuropathic pain

The gut microbiota plays a role in the maintenance of nervous system function, through immunological, hormonal, and neuronal signals.103103. Tognini P. Gut microbiota: a potential regulator of neurodevelopment. Front Cell Neurosci. 2017; 11: 25.,104104. Chen P, Wang C, Ren YN, Ye ZJ, Jiang C, Wu ZB. Alterations in the gut microbiota and metabolite profiles in the context of neuropathic pain. Mol Brain. 2021; 14(1): 50. Several studies showed the participation of gut microbiota in the development of pain, since GF mice exhibited visceral hypersensitivity that was controlled by postnatal colonization with conventional microbiota.105105. Sudo N, Chida Y, Aiba Y, Sonoda J, Oyama N, Yu XN, et al. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol. 2004; 558(Pt 1): 263-75.,106106. O' Mahony SM, Clarke G, McKernan DP, Bravo JA, Dinan TG, Cryan JF. Differential visceral nociceptive, behavioural and neurochemical responses to an immune challenge in the stress-sensitive Wistar Kyoto rat strain. Behav Brain Res. 2013; 253: 310-7.,107107. Luczynski P, Tramullas M, Viola M, Shanahan F, Clarke G, O'Mahony S, et al. Microbiota regulates visceral pain in the mouse. Elife. 2017; 6: e25887.,108108. Crouzet L, Gaultier E, Del'Homme C, Cartier C, Delmas E, Dapoigny M, et al. The hypersensitivity to colonic distension of IBS patients can be transferred to rats through their fecal microbiota. Neurogastroenterol Motil. 2013; 25(4): e272-82. In addition, the transference of fecal microbiota from irritable bowel syndrome patients to GF mice increased visceral hypersensitivity.109109. Distrutti E, Cipriani S, Mencarelli A, Renga B, Fiorucci S. Probiotics VSL#3 protect against development of visceral pain in murine model of irritable bowel syndrome. PLoS One. 2013; 8(5): e63893. Likewise, probiotic treatment improves stress- and inflammation-induced visceral hypersensitivity.110110. Eutamene H, Lamine F, Chabo C, Theodorou V, Rochat F, Bergonzelli GE, et al. Synergy between Lactobacillus paracasei and its bacterial products to counteract stress-induced gut permeability and sensitivity increase in rats. J Nutr. 2007; 137(8): 1901-7.,111111. Miquel S, Martín R, Lashermes A, Gillet M, Meleine M, et al. Anti-nociceptive effect of Faecalibacterium prausnitzii in non-inflammatory IBS-like models. Sci Rep. 2016; 6: 19399.,112112. Agostini S, Goubern M, Tondereau V, Salvador-Cartier C, Bezirard V, Lévèque M, et al. A marketed fermented dairy product containing Bifidobacterium lactis CNCM I-2494 suppresses gut hypersensitivity and colonic barrier disruption induced by acute stress in rats. Neurogastroenterol Motil. 2012; 24(4): 376-e172.,113113. Dai C, Guandalini S, Zhao DH, Jiang M. Antinociceptive effect of VSL#3 on visceral hypersensitivity in a rat model of irritable bowel syndrome: a possible action through nitric oxide pathway and enhance barrier function. Mol Cell Biochem. 2012; 362(1-2): 43-53. In addition, gut dysbiosis has been described in patients with irritable bowel syndrome who present abdominal pain.114114. Si JM, Yu YC, Fan YJ, Chen SJ. Intestinal microecology and quality of life in irritable bowel syndrome patients. World J Gastroenterol. 2004; 10(12): 1802-5.,115115. Rajilic-Stojanovic M, Biagi E, Heilig HG, Kajander K, Kekkonen RA, Tims S, et al. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology. 2011; 141(5): 1792-801.,116116. Pinn DM, Aroniadis OC, Brandt LJ. Is fecal microbiota transplantation the answer for irritable bowel syndrome? A single-center experience. Am J Gastroenterol. 2014; 109(11): 1831-2.,117117. Johnsen PH, Hilpüsch F, Cavanagh JP, Leikanger IS, Kolstad C, Valle PC, et al. Faecal microbiota transplantation versus placebo for moderate-to-severe irritable bowel syndrome: a double-blind, randomised, placebo-controlled, parallel-group, single-centre trial. Lancet Gastroenterol Hepatol. 2018; 3(1): 17-24.

Furthermore, the gut microbiota is also involved in the pathophysiology of neuropathic pain. For instance, some chemotherapy drugs, including paclitaxel and oxaliplatin, cause chemotherapy-induced peripheral pain (CIPN) during anti-cancer treatment,118118. Cavaletti G, Marmiroli P. Chemotherapy-induced peripheral neurotoxicity. Nat Rev Neurol. 2010; 6(12): 657-66. and affects up to 48% of patients undergoing chemotherapy.119119. Seretny M, Currie GL, Sena ES, Ramnarine S, Grant R, MacLeod MR, et al. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: a systematic review and meta-analysis. Pain. 2014; 155(12): 2461-70. Several chemotherapy drugs that induced neuropathic pain change the composition of gut microbiota, inducing dysbiosis.120120. Zhong S, Zhou Z, Liang Y, Cheng X, Li Y, Teng W, et al. Targeting strategies for chemotherapy-induced peripheral neuropathy: does gut microbiota play a role? Crit Rev Microbiol. 2019; 45(4): 369-93. Likewise, oxaliplatin-induced mechanical hyperalgesia is decreased in GF mice or in animals treated with antibiotics,121121. Shen S, Lim G, You Z, Ding W, Huang P, Ran C, et al. Gut microbiota is critical for the induction of chemotherapy-induced pain. Nat Neurosci. 2017; 20(9): 1213-6. suggesting that CIPN depends on dysbiosis. Another case of gut microbiota participating in neuropathic pain is observed in a murine model of chronic constriction injury (CCI).104104. Chen P, Wang C, Ren YN, Ye ZJ, Jiang C, Wu ZB. Alterations in the gut microbiota and metabolite profiles in the context of neuropathic pain. Mol Brain. 2021; 14(1): 50. In this model, the oral treatment with antibiotics resulted in gut microbiota changes and decreased the development of CCI-induced neuropathic pain, through a skewing from a pro-inflammatory to an anti-inflammatory immune profile,122122. Ding W, You Z, Chen Q, Yang L, Doheny J, Zhou X, et al. Gut microbiota influences neuropathic pain through modulating proinflammatory and anti-inflammatory T cells. Anesth Analg. 2021; 132(4): 1146-55. indicating that dysbiosis is an important factor in the development of CCI-induced neuropathic pain. In addition, the transplantation of fecal microbiota from rats with spared nerve injury to pseudo-GF mice increased mechanical stimulus-induced pain,123123. Yang C, Fang X, Zhan G, Huang N, Li S, Bi J, et al. Key role of gut microbiota in anhedonia-like phenotype in rodents with neuropathic pain. Transl Psychiatry. 2019; 9(1): 57 suggesting that gut microbiota possess a significant role in the spared nerve injury-induced neuropathic pain.

Since the development of neuropathic pain can be related to changes in the gut microbiota and diabetic patients presented dysbiosis in association with neuropathy, a central question arises: can dysbiosis and the consequent pro-inflammatory status be critical for the development of neuropathic pain in diabetes?

Dysbiosis is important to the establishment and aggravation of diabetic neuropathy

In diabetes, dysbiosis occurs in parallel to a break in the epithelial-intestinal barrier and translocation of bacterial contents to the bloodstream.6868. Durazzo M, Ferro A, Gruden G. Gastrointestinal microbiota and type 1 diabetes mellitus: the state of art. J Clin Med. 2019; 8(11): 1843. In addition, the hyperglycemia observed in diabetic patients is followed by an increase in the pro-inflammatory cytokines IL-1β, IL-6, and TNF-α, marking low-grade inflammation.7979. Popko K, Gorska E, Stelmaszczyk-Emmel A, Plywaczewski R, Stoklosa A, Gorecka D, et al. Proinflammatory cytokines Il-6 and TNF-a and the development of inflammation in obese subjects. Eur J Med Res. 2010; 15(Suppl. 2):120-2. Furthermore, the reduction in the butyrate-producing bacteria in the gut microbiota of diabetic patients is related to the high permeability of the epithelial-intestinal barrier.124124. de Groot PF, Belzer C, Aydin Ö, Levin E, Levels JH, Aalvink S, et al. Distinct fecal and oral microbiota composition in human type 1 diabetes, an observational study. PLoS One. 2017; 12(12): e0188475.

Interestingly, the incubation of Schwann cells in a high glucose environment induced an increase in the apoptosis of those cells in association with overexpression of TLR4, the receptor activated by LPS, and a rise in the TNF-α production.125125. Tang W, Chen X, Liu H, Lv Q, Zou J, Shi Y, et al. Expression of Nrf2 promotes Schwann cell-mediated sciatic nerve recovery in diabetic peripheral neuropathy. Cell Physiol Biochem. 2018; 46(5): 1879-94. In addition, the expression of TLR4 mRNA and the protein levels TNF-α were increased in the spinal cord of streptozotocin-induced diabetes. These raises in TLR4 and TNF-α were positively correlated with mechanical/thermal hypersensitivity in diabetic rats.126126. Yan JE, Yuan W, Lou X, Zhu T. Streptozotocin-induced diabetic hyperalgesia in rats is associated with upregulation of Toll-like receptor 4 expression. Neurosci Lett. 2012; 526(1): 54-8. Reinforcing the hypothesis that the increase in TLR4 expression in the Schwann cells and spinal cord is important to diabetic neuropathy, the inhibition of TLR4 signaling in the spinal cord attenuated mechanical hyperalgesia in diabetic rats with neuropathy and downregulated the local levels of TNF-α.127127. Liu P, Yuan HB, Zhao S, Liu FF, Jiang YQ, Guo YX, et al. Activation of GABAB receptor suppresses diabetic neuropathic pain through toll-like receptor 4 signaling pathway in the spinal dorsal horn. Mediators Inflamm. 2018; 2018: 6016272. Furthermore, the continuous delivery of IL-10 in the nerve fibers of DRG blocked the nociceptive response in diabetic animals, in parallel to a decrease in the expression of TLR4.128128. Thakur V, Gonzalez M, Pennington K, Chattopadhyay M. Viral vector mediated continuous expression of interleukin-10 in DRG alleviates pain in type 1 diabetic animals. Mol Cell Neurosci. 2016; 72: 46-53.

TNF-α is produced primarily by endoneurial macrophages and Schwann cells,129129. Wagner R, Myers RR. Schwann cells produce tumor necrosis factor alpha: expression in injured and non-injured nerves. Neuroscience. 1996; 73(3): 625-9. and is increased at the injury site after CCI of the sciatic nerve in rats.130130. George A, Schmidt C, Weishaupt A, Toyka KV, Sommer C. Serial determination of tumor necrosis factor-alpha content in rat sciatic nerve after chronic constriction injury. Exp Neurol. 1999; 160(1): 124-32.,131131. Shubayev VI, Myers RR. Upregulation and interaction of TNFalpha and gelatinases A and B in painful peripheral nerve injury. Brain Res. 2000; 855(1): 83-9. The administration of TNF-α inhibitor or antibodies to TNF-α reduced nerve injury- and CCI-induced hypersensitivity, respectively.132132. Sommer C, Schmidt C, George A. Hyperalgesia in experimental neuropathy is dependent on the TNF receptor 1. Exp Neurol. 1998; 151(1): 138-42.,133133. Iwatsuki K, Arai T, Ota H, Kato S, Natsume T, Kurimoto S, et al. Targeting anti-inflammatory treatment can ameliorate injury-induced neuropathic pain. PLoS One. 2013; 8(2): e57721. Furthermore, the IL1R1/TNFR1 double knock-out mice showed a decrease in the nociceptive sensitivity after nerve injury compared to wild-type littermates.134134. Nadeau S, Filali M, Zhang J, Kerr BJ, Rivest S, Soulet D, et al. Functional recovery after peripheral nerve injury is dependent on the pro-inflammatory cytokines IL-1ß and TNF: implications for neuropathic pain. J Neurosci. 2011; 31(35): 12533-42. In addition, direct injection of TNF-α into the sciatic nerve induced painful neuropathy.135135. Sorkin LS, Doom CM. Epineurial application of TNF elicits an acute mechanical hyperalgesia in the awake rat. J Peripher Nerv Syst. 2000; 5(2): 96-100.,136136. Wagner R, Myers RR. Endoneurial injection of TNF-alpha produces neuropathic pain behaviors. Neuroreport. 1996; 7(18): 2897-901. It is well known that patients with diabetic peripheral neuropathy showed elevated levels of both TNF-α and soluble TNF-α receptors in their serum.137137. Zoppini G, Faccini G, Muggeo M, Zenari L, Falezza G, Targher G. Elevated plasma levels of soluble receptors of TNF-alpha and their association with smoking and microvascular complications in young adults with type 1 diabetes. J Clin Endocrinol Metab. 2001; 86(8): 3805-8.,138138. González-Clemente JM, Mauricio D, Richart C, Broch M, Caixàs A, Megia A, et al. Diabetic neuropathy is associated with activation of the TNF-alpha system in subjects with type 1 diabetes mellitus. Clin Endocrinol (Oxf). 2005; 63(5): 525-9.,139139. Moriwaki Y, Yamamoto T, Shibutani Y, Aoki E, Tsutsumi Z, Takahashi S, et al. Elevated levels of interleukin-18 and tumor necrosis factor-alpha in serum of patients with type 2 diabetes mellitus: relationship with diabetic nephropathy. Metabolism. 2003; 52(5): 605-8. The inhibition of TNF-α, using a recombinant human TNF-α receptor-antibody fusion protein, recovery lower nerve conduction velocity, demyelination of nerve fibers, disorganization of lamellar and axonal structures, and decreased expression of myelin basic protein in the nerve tissue of diabetic rats that developed peripheral neuropathy.140140. Shi X, Chen Y, Nadeem L, Xu G. Beneficial effect of TNF-a inhibition on diabetic peripheral neuropathy. J Neuroinflammation. 2013; 10: 69. Furthermore, the blockage of TNF-α signaling using either TNF-α knockout mice or anti-TNF-α monoclonal antibody improved neuropathy in diabetic mice.141141. Yamakawa I, Kojima H, Terashima T, Katagi M, Oi J, Urabe H, et al. Inactivation of TNF-a ameliorates diabetic neuropathy in mice. Am J Physiol Endocrinol Metab. 2011; 301(5): E844-52.

Sodium butyrate has anti-inflammatory and neuroprotective effects in spinal cord injury, including the improvement of motor function and the reduction of neutrophils accumulation and pro-inflammatory cytokine expression.142142. Lanza M, Campolo M, Casili G, Filippone A, Paterniti I, Cuzzocrea S, et al. Sodium butyrate exerts neuroprotective effects in spinal cord injury. Mol Neurobiol. 2019; 56(6): 3937-47. Furthermore, sodium butyrate and sodium propionate improved nitroglycerin-induced pain attacks, reducing the damage in the trigeminal nerve nucleus and the expression of pro-inflammatory mediators.143143. Lanza M, Filippone A, Ardizzone A, Casili G, Paterniti I, Esposito E, et al. SCFA Treatment alleviates pathological signs of migraine and related intestinal alterations in a mouse model of NTG-induced migraine. Cells. 2021; 10(10): 2756. Likewise, the pain and discomfort in healthy human were reduced after intraluminal administration of butyrate into the distal colon.144144. Vanhoutvin SA, Troost FJ, Kilkens TO, Lindsey PJ, Hamer HM, Jonkers DM, et al. The effects of butyrate enemas on visceral perception in healthy volunteers. Neurogastroenterol Motil. 2009; 21(9): 952-e76. Altogether, these evidences suggested that the decrease in the butyrate-producing bacteria in diabetic patients can be related to the development of neuropathic pain.

The composition of the gut microbiota can change during the development of diabetes. Interestingly, the diversity of gut microbiota from patients with type 2 diabetes with gastrointestinal autonomic neuropathy was modified compared to type 2 diabetic patients without this condition. Diabetic patients with neuropathy presented an increase in the relative abundance of pathogenic bacteria of phyla Proteobacteria,145145. Du Y, Neng Q, Li Y, Kang Y, Guo L, Huang X, et al. Gastrointestinal autonomic neuropathy exacerbates gut microbiota dysbiosis in adult patients with type 2 diabetes mellitus. Front Cell Infect Microbiol. 2022; 11: 804733. a LPS-producing bacteria phylum.146146. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol. 2009; 9(5): 313-23. Jinmaitong, a natural compound rich in flavonoid and its glycosides, triterpenoids, and phenolic acids,147147. Song W, Jiang W, Wang C, Xie J, Liang X, Sun Y, et al. Jinmaitong, a traditional Chinese compound prescription, ameliorates the streptozocin-induced diabetic peripheral neuropathy rats by increasing sciatic nerve IGF-1 and IGF-1R expression. Front Pharmacol. 2019; 10: 255. improves nerve conduction velocity, pain, and temperature sensation in diabetic rats with neuropathy,148148. Shi Y, Liang XC, Wu QL, Sun LQ, Qu L, Zhao L, et al. Effects of Jinmaitong capsule on ciliary neurotrophic factor in sciatic nerves of diabetes mellitus rats. Chin J Integr Med. 2013; 19(2): 104-11.,149149. Yin DH, Liang XC, Zhao LI, Zhang H, Sun Q, Wang PY, et al. Jinmaitong decreases sciatic nerve DNA oxidative damage and apoptosis in a streptozotocin-induced diabetic rat model. Exp Ther Med. 2015; 10(2): 778-86. as well as markedly ameliorated clinical symptoms of pain in the extremities of diabetic patients with peripheral neuropathy.150150. Liang X, Cui L, Guo S. Clinical study on jinmaitong composita on diabetic peripheral neuropathy. Zhongguo Zhong. Xi Yi Jie He Za Zhi. 1999; 19(9): 517-9. In parallel, Jinmaitong enriched nine species of gut microbiota of diabetic rats with neuropathy, avoiding dysbiosis.151151. Xie J, Song W, Liang X, Zhang Q, Shi Y, Liu W, et al. Jinmaitong ameliorates diabetic peripheral neuropathy in streptozotocin-induced diabetic rats by modulating gut microbiota and neuregulin 1. Aging (Albany NY). 2020; 12(17): 17436-58.

Furthermore, the flavonoid quercetin reversed mechanical pain and intraepidermal nerve fiber density in streptozotocin-induced diabetic rats, in clear association with the reduction of pathogenic bacteria species and the enrichment of two prebiotic species.152152. Xie J, Song W, Liang X, Zhang Q, Shi Y, Liu W, et al. Protective effect of quercetin on streptozotocin-induced diabetic peripheral neuropathy rats through modulating gut microbiota and reactive oxygen species level. Biomed Pharmacother. 2020; 127: 110147. These data suggest that both Jinmaitong and quercetin improve neuropathy in diabetic subjects by modulating phenotype-associated gut microbiota. In agreement with the proposition that dysbiosis can be important to the development of neuropathy in diabetes, a case report showed that fecal microbiota transplantation decrease limb pain and paresthesia in a diabetic patient with neuropathy that did not use any painkillers or drugs for alleviating the pain. In addition, this patient showed an improvement of motor conduction velocity in tibial nerve, attested by electromyogram, and a reduction in the visual analogue scale pain score from severe pain to mild pain after the treatment with fecal microbiota transplantation.153153. Cai TT, Ye XL, Yong HJ, Song B, Zheng XL, Cui BT, et al. Fecal microbiota transplantation relieve painful diabetic neuropathy: a case report. Medicine (Baltimore). 2018; 97(50): e13543.

In conclusion

In conclusion, we postulate that the increase in the richness of pathogenic bacteria and a reduction in the abundance of butyrate-producing bacteria in the gut microbiota of diabetic patients may be responsible for the onset of peripheral neuropathy. The dysbiosis in diabetes triggers a break in the intestinal barrier with consequent increase in the bacterial products, such as LPS, into the bloodstream. Possibly, the activation of TLR4 in the Schwann cells and spinal cord of diabetics induces an overproduction of TNF-α, resulting in the increase of pain (Fig. 2). In this respect, new therapeutic strategies founded on probiotics or bacterial metabolites, as butyrate, seem to be potentially practical approaches for adjuvant treatment of neuropathy in diabetic patients.

Fig. 2:
mechanisms associated with diabetic neuropathy induced by gut microbiota dysbiosis and the therapeutic effects of prebiotics/probiotics. Dysbiosis in diabetic patients results in a leak of epithelial-intestinal barrier, increasing its permeability and the absorption of molecules from lumen to the blood such as lipopolysaccharide (LPS). LPS directly activates Schwann cells to release tumor necrosis factor-α (TNF-α), favoring the nociception in diabetics. Changes in diet, prebiotics or probiotics may induces changes in microbiota, restoring butyrate producing microorganisms, leading to anti-inflammatory effects, reducing the TNF-α production and, consequently, the pain in diabetics tumor necrosis factor.

REFERENCES

  • 1
    Insuela DBR, Ferrero MR, Gonçalves-de-Albuquerque CF, Chaves ADS, da Silva AYO, Castro-Faria-Neto HC, et al. Glucagon reduces neutrophil migration and increases susceptibility to sepsis in diabetic mice. Front Immunol. 2021; 12: 633540.
  • 2
    Heald AH, Stedman M, Davies M, Livingston M, Alshames R, Lunt M, et al. Estimating life years lost to diabetes: outcomes from analysis of National Diabetes Audit and Office of National Statistics data. Cardiovasc Endocrinol Metab. 2020; 9(4): 183-5.
  • 3
    Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al. IDF diabetes atlas: global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract. 2022; 183: 109119.
  • 4
    Feldman EL, Callaghan BC, Pop-Busui R, Zochodne DW, Wright DE, Bennett DL, et al. Diabetic neuropathy. Nat Rev Dis Primers. 2019; 5(1): 41.
  • 5
    Patel S, Farkash C, Simmons D. Type 1 diabetes management and hospitalisation in the over 25's at an Australian outer urban diabetes clinic. BMC Endocr Disord. 2022; 22(1): 143.
  • 6
    Hicks CW, Selvin E. Epidemiology of peripheral neuropathy and lower extremity disease in diabetes. Curr Diab Rep. 2019; 19(10): 86.
  • 7
    Kamerman PR, Wadley AL, Davis KD, Hietaharju A, Jain P, Kopf A, et al. World Health Organization essential medicines lists: where are the drugs to treat neuropathic pain? Pain. 2015; 156(5): 793-7.
  • 8
    Finnerup NB, Attal N, Haroutounian S, McNicol E, Baron R, Dworkin RH, et al. Pharmacotherapy for neuropathic pain in adults: a systematic review and meta-analysis. Lancet Neurol. 2015; 14(2): 162-73.
  • 9
    Scott JN, Clark AW, Zochodne DW. Neurofilament and tubulin gene expression in progressive experimental diabetes: failure of synthesis and export by sensory neurons. Brain. 1999; 122(11): 2109-18.
  • 10
    Ma J, Pan P, Anyika M, Blagg BS, Dobrowsky RT. Modulating molecular chaperones improves mitochondrial bioenergetics and decreases the inflammatory transcriptome in diabetic sensory neurons. ACS Chem Neurosci. 2015; 6(9): 1637-48.
  • 11
    Urban MJ, Pan P, Farmer KL, Zhao H, Blagg BS, Dobrowsky RT. Modulating molecular chaperones improves sensory fiber recovery and mitochondrial function in diabetic peripheral neuropathy. Exp Neurol. 2012; 235(1): 388-96.
  • 12
    Ilnytska O, Lyzogubov VV, Stevens MJ, Drel VR, Mashtalir N, Pacher P, et al. Poly(ADP-ribose) polymerase inhibition alleviates experimental diabetic sensory neuropathy. Diabetes. 2006; 55(6): 1686-94.
  • 13
    Lupachyk S, Shevalye H, Maksimchyk Y, Drel VR, Obrosova IG. PARP inhibition alleviates diabetes-induced systemic oxidative stress and neural tissue 4-hydroxynonenal adduct accumulation: correlation with peripheral nerve function. Free Radic Biol Med. 2011; 50(10): 1400-9.
  • 14
    Cashman CR, Höke A. Mechanisms of distal axonal degeneration in peripheral neuropathies. Neurosci Lett. 2015; 596: 33-50.
  • 15
    Feldman EL, Nave KA, Jensen TS, Bennett DLH. New horizons in diabetic neuropathy: mechanisms, bioenergetics, and pain. Neuron. 2017; 93(6): 1296-1313.
  • 16
    Dunnigan SK, Ebadi H, Breiner A, Katzberg HD, Lovblom LE, Perkins BA, et al. Conduction slowing in diabetic sensorimotor polyneuropathy. Diabetes Care. 2013; 36(11): 3684-90.
  • 17
    Gumy LF, Bampton ET, Tolkovsky AM. Hyperglycaemia inhibits Schwann cell proliferation and migration and restricts regeneration of axons and Schwann cells from adult murine DRG. Mol Cell Neurosci. 2008; 37(2): 298-311.
  • 18
    Mizisin AP, Shelton GD, Wagner S, Rusbridge C, Powell HC. Myelin splitting, Schwann cell injury and demyelination in feline diabetic neuropathy. Acta Neuropathol. 1998; 95(2): 171-4.
  • 19
    Zochodne DW, Ho LT. The influence of sulindac on experimental streptozotocin-induced diabetic neuropathy. Can J Neurol Sci. 1994; 21(3): 194-202.
  • 20
    Kang Q, Yang C. Oxidative stress and diabetic retinopathy: molecular mechanisms, pathogenetic role and therapeutic implications. Redox Biol. 2020; 37: 101799.
  • 21
    Jang ER, Lee CS. 7-ketocholesterol induces apoptosis in differentiated PC12 cells via reactive oxygen species-dependent activation of NF-?B and Akt pathways. Neurochem Int. 2011; 58(1): 52-9.
  • 22
    Legrand-Poels S, Esser N, L'homme L, Scheen A, Paquot N, Piette J. Free fatty acids as modulators of the NLRP3 inflammasome in obesity/type 2 diabetes. Biochem Pharmacol. 2014; 92(1): 131-41.
  • 23
    Burkart V, Wang ZQ, Radons J, Heller B, Herceg Z, Stingl L, et al. Mice lacking the poly(ADP-ribose) polymerase gene are resistant to pancreatic beta-cell destruction and diabetes development induced by streptozocin. Nat Med. 1999; 5(3): 314-9.
  • 24
    Fernyhough P, McGavock J. Mechanisms of disease: mitochondrial dysfunction in sensory neuropathy and other complications in diabetes. Handb Clin Neurol. 2014; 126: 353-77.
  • 25
    Chowdhury SK, Smith DR, Fernyhough P. The role of aberrant mitochondrial bioenergetics in diabetic neuropathy. Neurobiol Dis. 2013; 51: 56-65.
  • 26
    Huvers FC, De Leeuw PW, Houben AJ, De Haan CH, Hamulyak K, Schouten H, et al. Endothelium-dependent vasodilatation, plasma markers of endothelial function, and adrenergic vasoconstrictor responses in type 1 diabetes under near-normoglycemic conditions. Diabetes. 1999; 48(6): 1300-7.
  • 27
    Rumora AE, Lentz SI, Hinder LM, Jackson SW, Valesano A, Levinson GE, et al. Dyslipidemia impairs mitochondrial trafficking and function in sensory neurons. FASEB J. 2018; 32(1): 195-207.
  • 28
    Hink U, Li H, Mollnau H, Oelze M, Matheis E, Hartmann M, et al. Mechanisms underlying endothelial dysfunction in diabetes mellitus. Circ Res. 2001; 88(2): e14-e22.
  • 29
    Vinik A, Ullal J, Parson HK, Casellini CM. Diabetic neuropathies: clinical manifestations and current treatment options. Nat Clin Pract Endocrinol Metab. 2006; 2(5): 269-81.
  • 30
    Kasznicki J, Kosmalski M, Sliwinska A, Mrowicka M, Stanczyk M, Majsterek I, et al. Evaluation of oxidative stress markers in pathogenesis of diabetic neuropathy. Mol Biol Rep. 2012; 39(9): 8669-78.
  • 31
    Hur J, O'Brien PD, Nair V, Hinder LM, McGregor BA, Jagadish HV, et al. Transcriptional networks of murine diabetic peripheral neuropathy and nephropathy: common and distinct gene expression patterns. Diabetologia. 2016; 59(6): 1297-306.
  • 32
    McGregor BA, Eid S, Rumora AE, Murdock B, Guo K, de Anda-Jáuregui G, et al. Conserved transcriptional signatures in human and murine diabetic peripheral neuropathy. Sci Rep. 2018; 8(1): 17678.
  • 33
    Hur J, Sullivan KA, Pande M, Hong Y, Sima AA, Jagadish HV, et al. The identification of gene expression profiles associated with progression of human diabetic neuropathy. Brain. 2011; 134(Pt 11): 3222-35.
  • 34
    Hur J, Sullivan KA, Callaghan BC, Pop-Busui R, Feldman EL. Identification of factors associated with sural nerve regeneration and degeneration in diabetic neuropathy. Diabetes Care. 2013; 36(12): 4043-9.
  • 35
    Baum P, Kosacka J, Estrela-Lopis I, Woidt K, Serke H, Paeschke S, et al. The role of nerve inflammation and exogenous iron load in experimental peripheral diabetic neuropathy (PDN). Metabolism. 2016; 65(4): 391-405.
  • 36
    Kosacka J, Woidt K, Toyka KV, Paeschke S, Klöting N, Bechmann I, et al. The role of dietary non-heme iron load and peripheral nerve inflammation in the development of peripheral neuropathy (PN) in obese non-diabetic leptin-deficient ob/ob mice. Neurol Res. 2019; 41(4): 341-53.
  • 37
    Paeschke S, Baum P, Toyka KV, Blüher M, Koj S, Klöting N, et al. The role of iron and nerve inflammation in diabetes mellitus type 2-induced peripheral neuropathy. Neuroscience. 2019; 406: 496-509.
  • 38
    Cheng HT, Dauch JR, Oh SS, Hayes JM, Hong Y, Feldman EL. p38 mediates mechanical allodynia in a mouse model of type 2 diabetes. Mol Pain. 2010; 6: 28.
  • 39
    Kosacka J, Nowicki M, Klöting N, Kern M, Stumvoll M, Bechmann I, et al. COMP-angiopoietin-1 recovers molecular biomarkers of neuropathy and improves vascularisation in sciatic nerve of ob/ob mice. PLoS One. 2012; 7(3): e32881.
  • 40
    Herder C, Kannenberg JM, Huth C, Carstensen-Kirberg M, Rathmann W, Koenig W, et al. Proinflammatory cytokines predict the incidence and progression of distal sensorimotor polyneuropathy: KORA F4/FF4 study. Diabetes Care. 2017; 40(4): 569-76.
  • 41
    Hall BE, Macdonald E, Cassidy M, Yun S, Sapio MR, Ray P, et al. Transcriptomic analysis of human sensory neurons in painful diabetic neuropathy reveals inflammation and neuronal loss. Sci Rep. 2022; 12(1): 4729.
  • 42
    Chowdhury SK, Zherebitskaya E, Smith DR, Akude E, Chattopadhyay S, Jolivalt CG, et al. Mitochondrial respiratory chain dysfunction in dorsal root ganglia of streptozotocin-induced diabetic rats and its correction by insulin treatment. Diabetes. 2010; 59(4): 1082-91.
  • 43
    Fernyhough P, Roy Chowdhury SK, Schmidt RE. Mitochondrial stress and the pathogenesis of diabetic neuropathy. Expert Rev Endocrinol Metab. 2010; 5(1): 39-49.
  • 44
    Ma J, Farmer KL, Pan P, Urban MJ, Zhao H, Blagg BS, et al. Heat shock protein 70 is necessary to improve mitochondrial bioenergetics and reverse diabetic sensory neuropathy following KU-32 therapy. J Pharmacol Exp Ther. 2014; 348(2): 281-92.
  • 45
    Ma J, Pan P, Anyika M, Blagg BS, Dobrowsky RT. Modulating molecular chaperones improves mitochondrial bioenergetics and decreases the inflammatory transcriptome in diabetic sensory neurons. ACS Chem Neurosci. 2015; 6(9): 1637-48.
  • 46
    Bäckryd E, Themistocleous A, Larsson A, Gordh T, Rice AS, Tesfaye S, et al. Hepatocyte growth factor, colony-stimulating factor 1, CD40, and 11 other inflammation-related proteins are associated with pain in diabetic neuropathy: exploration and replication serum data from the Pain in Neuropathy Study. Pain. 2022; 163(5): 897-909.
  • 47
    Thursby E, Juge N. Introduction to the human gut microbiota. Biochem J. 2017; 474(11): 1823-36.
  • 48
    Vemuri R, Shankar EM, Chieppa M, Eri R, Kavanagh K. Beyond just bacteria: functional biomes in the gut ecosystem including virome, mycobiome, archaeome and helminths. Microorganisms. 2020; 8(4): 483.
  • 49
    Tsang DK, Wang RJ, De Sa O, Ayyaz A, Foerster EG, Bayer G, et al. A single cell survey of the microbial impacts on the mouse small intestinal epithelium. Gut Microbes. 2022; 14(1): 2108281.
  • 50
    Gao Z, Yin J, Zhang J, Ward RE, Martin RJ, Lefevre M, et al. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes. 2009; 58(7): 1509-17.
  • 51
    Edwinson A, Yang L, Chen J, Grover, M. Colonic expression of Ace2, the SARS-CoV-2 entry receptor, is suppressed by commensal human microbiota. Gut Microbes. 2021; 13(1): 1984105.
  • 52
    Romero R, Zarzycka A, Preussner M, Fischer F, Hain T, Herrmann JP, et al. Selected commensals educate the intestinal vascular and immune system for immunocompetence. Microbiome. 2022; 10(1): 1-18.
  • 53
    Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. Human nutrition, the gut microbiome and the immune system. Nature. 2011; 474(7351): 327-36.
  • 54
    Pérez-Cobas AE, Gosalbes MJ, Friedrichs A, Knecht H, Artacho A, Eismann K, et al. Gut microbiota disturbance during antibiotic therapy: a multi-omic approach. Gut. 2013; 62(11): 1591-1601.
  • 55
    Levy M, Kolodziejczyk AA, Thaiss CA, Elinav E. Dysbiosis and the immune system. Nat Rev Immunol. 2017; 17(4): 219-232.
  • 56
    Weiss GA, Hennet T. Mechanisms and consequences of intestinal dysbiosis. Cell Mol Life Sci. 2017; 74(16): 2959-77.
  • 57
    Weintraub AS, Ferrara L, Deluca L, Moshier E, Green RS, Oakman E, et al. Antenatal antibiotic exposure in preterm infants with necrotizing enterocolitis. J Perinatol. 2012; 32(9): 705-9.
  • 58
    Cotten CM, Taylor S, Stoll B, Goldberg RN, Hansen NI, Sánchez PJ, et al. Prolonged duration of initial empirical antibiotic treatment is associated with increased rates of necrotizing enterocolitis and death for extremely low birth weight infants. Pediatrics. 2009; 123(1): 58-66.
  • 59
    Kim JE, Li B, Fei L, Horne R, Lee D, Loe AK, et al. Gut microbiota promotes stem cell differentiation through macrophage and mesenchymal niches in early postnatal development. Immunity. 2022; 55(12): 2300-17.
  • 60
    Li P, Chang X, Chen X, Wang C, Shang Y, Zheng D, et al. Early-life antibiotic exposure increases the risk of childhood overweight and obesity in relation to dysbiosis of gut microbiota: a birth cohort study. Ann Clin Microbiol Antimicrob. 2022; 21(1): 46.
  • 61
    Ruiz-Ojeda FJ, Plaza-Díaz J, Sáez-Lara MJ, Gil A. Effects of sweeteners on the gut microbiota: a review of experimental studies and clinical trials. Adv Nutr. 2019; 1(10): S31-48.
  • 62
    Li CH, Wang CT, Lin YJ, Kuo HY, Wu JS, Hong TC, et al. Long-term consumption of the sugar substitute sorbitol alters gut microbiome and induces glucose intolerance in mice. Life Sci. 2022; 305: 120770.
  • 63
    Alam C, Bittoun E, Bhagwat D, Valkonen S, Saari A, Jaakkola U, et al. Effects of a germ-free environment on gut immune regulation and diabetes progression in non-obese diabetic (NOD) mice. Diabetologia. 2011; 54(6): 1398-406.
  • 64
    King C, Sarvetnick N. The incidence of type-1 diabetes in NOD mice is modulated by restricted flora not germ-free conditions. PLoS One. 2011; 6(2): e17049.
  • 65
    Calcinaro F, Dionisi S, Marinaro M, Candeloro P, Bonato V, et al. Oral probiotic administration induces interleukin-10 production and prevents spontaneous autoimmune diabetes in the non-obese diabetic mouse. Diabetologia. 2005; 48(8): 1565-75.
  • 66
    Matsuzaki T, Nagata Y, Kado S, Uchida K, Kato I, Hashimoto S, et al. Prevention of onset in an insulin-dependent diabetes mellitus model, NOD mice, by oral feeding of Lactobacillus casei. APMIS. 1997; 105(8): 643-9.
  • 67
    Kim TK, Lee JC, Im SH, Lee MS. Amelioration of autoimmune diabetes of NOD mice by immunomodulating probiotics. Front Immunol. 2020; 11: 1832.
  • 68
    Durazzo M, Ferro A, Gruden G. Gastrointestinal microbiota and type 1 diabetes mellitus: the state of art. J Clin Med. 2019; 8(11): 1843.
  • 69
    Muñoz-Garach A, Diaz-Perdigones C, Tinahones FJ. Gut microbiota and type 2 diabetes mellitus. Endocrinol Nutr. 2016; 63(10): 560-8.
  • 70
    Santos-Marcos JA, Perez-Jimenez F, Camargo A. The role of diet and intestinal microbiota in the development of metabolic syndrome. J Nutr Biochem. 2019; 70: 1-27.
  • 71
    Ahmad A, Yang W, Chen G, Shafiq M, Javed S, Ali Zaidi SS, et al. Analysis of gut microbiota of obese individuals with type 2 diabetes and healthy individuals. PLoS One. 2019; 14(12): e0226372.
  • 72
    Pascale A, Marchesi N, Govoni S, Coppola A, Gazzaruso C. The role of gut microbiota in obesity, diabetes mellitus, and effect of metformin: new insights into old diseases. Curr Opin Pharmacol. 2019; 49: 1-5.
  • 73
    van Heck JIP, Gacesa R, Stienstra R, Fu J, Zhernakova A, Harmsen HJM, et al. The gut microbiome composition is altered in long-standing type 1 diabetes and associates with glycemic control and disease-related complications. Diabetes Care. 2022; dc212225.
  • 74
    Snelson M, de Pasquale C, Ekinci EI, Coughlan MT. Gut microbiome, prebiotics, intestinal permeability and diabetes complications. Best Pract Res Clin Endocrinol Metab. 2021; 35(3): 101507.
  • 75
    Meddings JB, Jarand J, Urbanski SJ, Hardin J, Gall DG. Increased gastrointestinal permeability is an early lesion in the spontaneously diabetic BB rat. Am J Physiol. 1999; 276(4): G951-7.
  • 76
    Carratù R, Secondulfo M, de Magistris L, Iafusco D, Urio A, Carbone MG, et al. Altered intestinal permeability to mannitol in diabetes mellitus type I. J Pediatr Gastroenterol Nutr. 1999; 28(3): 264-9.
  • 77
    Jayashree B, Bibin YS, Prabhu D, Shanthirani CS, Gokulakrishnan K, Lakshmi BS, et al. Increased circulatory levels of lipopolysaccharide (LPS) and zonulin signify novel biomarkers of proinflammation in patients with type 2 diabetes. Mol Cell Biochem. 2014; 388(1-2): 203-10.
  • 78
    Xu J, Liang R, Zhang W, Tian K, Li J, Chen X, et al. Faecalibacterium prausnitzii -derived microbial anti-inflammatory molecule regulates intestinal integrity in diabetes mellitus mice via modulating tight junction protein expression. J Diabetes. 2020; 12(3): 224-36.
  • 79
    Popko K, Gorska E, Stelmaszczyk-Emmel A, Plywaczewski R, Stoklosa A, Gorecka D, et al. Proinflammatory cytokines Il-6 and TNF-a and the development of inflammation in obese subjects. Eur J Med Res. 2010; 15(Suppl. 2):120-2.
  • 80
    Ejtahed HS, Hoseini-Tavassol Z, Khatami S, Zangeneh M, Behrouzi A, Ahmadi Badi S, et al. Main gut bacterial composition differs between patients with type 1 and type 2 diabetes and non-diabetic adults. J Diabetes Metab Disord. 2020; 19(1): 265-71.
  • 81
    Ganesan K, Chung SK, Vanamala J, Xu B. Causal relationship between diet-induced gut microbiota changes and diabetes: a novel strategy to transplant Faecalibacterium prausnitzii in preventing diabetes. Int J Mol Sci. 2018; 19(12): 3720.
  • 82
    Guilloteau P, Martin L, Eeckhaut V, Ducatelle R, Zabielski R, Van Immerseel F. From the gut to the peripheral tissues: the multiple effects of butyrate. Nutr Res Rev. 2010; 23(2): 366-84.
  • 83
    Khan S, Jena G. The role of butyrate, a histone deacetylase inhibitor in diabetes mellitus: experimental evidence for therapeutic intervention. Epigenomics. 2015; 7(4): 669-80.
  • 84
    Noureldein MH, Bitar S, Youssef N, Azar S, Eid AA. Butyrate modulates diabetes-linked gut dysbiosis: epigenetic and mechanistic modifications. J Mol Endocrinol. 2020; 64(1): 29-42.
  • 85
    Andreasen AS, Larsen N, Pedersen-Skovsgaard T, Berg RM, Møller K, Svendsen KD, et al. Effects of Lactobacillus acidophilus NCFM on insulin sensitivity and the systemic inflammatory response in human subjects. Br J Nutr. 2010; 104(12): 1831-8.
  • 86
    Yao K, Zeng L, He Q, Wang W, Lei J, Zou X. Effect of probiotics on glucose and lipid metabolism in type 2 diabetes mellitus: a meta-analysis of 12 randomized controlled trials. Med Sci Monit. 2017; 23: 3044-53.
  • 87
    Alfa MJ, Strang D, Tappia PS, Olson N, DeGagne P, Bray D, et al. A randomized placebo controlled clinical trial to determine the impact of digestion resistant starch MSPrebiotic(r) on glucose, insulin, and insulin resistance in elderly and mid-age adults. Front Med (Lausanne). 2018; 4: 260.
  • 88
    FAO/WHO - Food and Agriculture Organization/World Health Organization. Probiotics in food. Health and nutritional properties and guidelines for evaluation. Fao Food and Nutrition. 2006; paper 85. Available from: https://www.fao.org/3/a0512e/a0512e.pdf
    » https://www.fao.org/3/a0512e/a0512e.pdf
  • 89
    Wang Y, Liu H, Zheng M, Yang Y, Ren H, Kong Y, et al. Berberine slows the progression of prediabetes to diabetes in zucker diabetic fatty rats by enhancing intestinal secretion of glucagon-like peptide-2 and improving the gut microbiota. Front Endocrinol (Lausanne). 2021; 12: 609134.
  • 90
    Singh S, Sharma RK, Malhotra S, Pothuraju R, Shandilya UK. Lactobacillus rhamnosus NCDC17 ameliorates type-2 diabetes by improving gut function, oxidative stress and inflammation in high-fat-diet fed and streptozotocintreated rats. Benef Microbes. 2017; 8(2): 243-55.
  • 91
    Andreasen A, Larsen N, Pedersen-Skovsgaard T, Berg R, Møller K, Svendsen KD, et al. Effects of Lactobacillus acidophilus NCFM on insulin sensitivity and the systemic inflammatory response in human subjects. British J Nutr. 2010; 104: 1831-8.
  • 92
    Asemi Z, Zare Z, Shakeri H, Sabihi SS, Esmaillzadeh A. Effect of multispecies probiotic supplements on metabolic profiles, hs-CRP, and oxidative stress in patients with type 2 diabetes. Ann Nutr Metab. 2013; 63(1-2): 1-9.
  • 93
    Asemi Z, Khorrami-Rad A, Alizadeh SA, Shakeri H, Esmaillzadeh A. Effects of synbiotic food consumption on metabolic status of diabetic patients: a double-blind randomized cross-over controlled clinical trial. Clin Nutr. 2014; 33(2): 198-203.
  • 94
    Moroti C, Magri LFS, Costa MR, Cavallini DCU, Sivieri K. Effect of the consumption of a new symbiotic shake on glycemia and cholesterol levels in elderly people with type 2 diabetes mellitus. Lipids Health Dis. 2012; 11: 29.
  • 95
    Tonucci LB, Dos Santos KMO, Oliveira LL, Ribeiro SMR, Martino HSD. Clinical application of probiotics in type 2 diabetes mellitus: a randomized, double-blind, placebo-controlled study. Clin Nutr. 2017; 36(1): 85-92.
  • 96
    Sato J, Kanazawa A, Azuma K, Ikeda F, Goto H, Komiya K, et al. Probiotic reduces bacterial translocation in type 2 diabetes mellitus: a randomised controlled study. Sci Rep. 2017; 7(1): 1-10.
  • 97
    Ejtahed HS, Mohtadi-Nia J, Homayouni-Rad A, Niafar M, Asghari-Jafarabadi M, Mofid V. Probiotic yogurt improves probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition. 2012; 28(5): 539-43.
  • 98
    Kanazawa A, Aida M, Yoshida Y, Kaga H, Katahira T, Suzuki L, et al. Effects of synbiotic probiotication on chronic inflammation and the gut microbiota in obese patients with type 2 diabetes mellitus: a randomized controlled study. Nutrients. 2021; 13: 558.
  • 99
    Razmpoosh E, Javadi A, Ejtahed HS, Mirmiran P, Javadi M, Yousefinejad A. The effect of probiotic supplementation on glycemic control and lipid profile in patients with type 2 diabetes: a randomized placebo controlled trial. Diabetes Metab Syndr. 2019; 13(1): 175-82.
  • 100
    Khalili L, Alipour B, Jafar-Abadi MA, Faraji I, Hassanalilou T, Abbasi MM. The effects of Lactobacillus casei on glycemic response, serum sirtuin1 and fetuin-a levels in patients with type 2 diabetes mellitus: a randomized controlled trial. Iran Biomed J. 2019; 23(1): 68.
  • 101
    Kobyliak N, Falalyeyeva T, Mykhalchyshyn G, Kyriienko D, Komissarenko I. Effect of alive probiotic on insulin resistance in type 2 diabetes patients: randomized clinical trial. Diabetes Metab Syndr. 2018; 12(5): 617-24.
  • 102
    Wang CH, Yen HR, Lu WL, Ho HH, Lin WY, Kuo YW, et al. Adjuvant probiotics of Lactobacillus salivarius subsp. salicinius AP-32, L. johnsonii MH-68, and Bifidobacterium animalis subsp. lactis CP-9 attenuate glycemic levels and inflammatory cytokines in patients with type 1 diabetes mellitus. Front Endocrinol (Lausanne). 2022; 13: 754401.
  • 103
    Tognini P. Gut microbiota: a potential regulator of neurodevelopment. Front Cell Neurosci. 2017; 11: 25.
  • 104
    Chen P, Wang C, Ren YN, Ye ZJ, Jiang C, Wu ZB. Alterations in the gut microbiota and metabolite profiles in the context of neuropathic pain. Mol Brain. 2021; 14(1): 50.
  • 105
    Sudo N, Chida Y, Aiba Y, Sonoda J, Oyama N, Yu XN, et al. Postnatal microbial colonization programs the hypothalamic-pituitary-adrenal system for stress response in mice. J Physiol. 2004; 558(Pt 1): 263-75.
  • 106
    O' Mahony SM, Clarke G, McKernan DP, Bravo JA, Dinan TG, Cryan JF. Differential visceral nociceptive, behavioural and neurochemical responses to an immune challenge in the stress-sensitive Wistar Kyoto rat strain. Behav Brain Res. 2013; 253: 310-7.
  • 107
    Luczynski P, Tramullas M, Viola M, Shanahan F, Clarke G, O'Mahony S, et al. Microbiota regulates visceral pain in the mouse. Elife. 2017; 6: e25887.
  • 108
    Crouzet L, Gaultier E, Del'Homme C, Cartier C, Delmas E, Dapoigny M, et al. The hypersensitivity to colonic distension of IBS patients can be transferred to rats through their fecal microbiota. Neurogastroenterol Motil. 2013; 25(4): e272-82.
  • 109
    Distrutti E, Cipriani S, Mencarelli A, Renga B, Fiorucci S. Probiotics VSL#3 protect against development of visceral pain in murine model of irritable bowel syndrome. PLoS One. 2013; 8(5): e63893.
  • 110
    Eutamene H, Lamine F, Chabo C, Theodorou V, Rochat F, Bergonzelli GE, et al. Synergy between Lactobacillus paracasei and its bacterial products to counteract stress-induced gut permeability and sensitivity increase in rats. J Nutr. 2007; 137(8): 1901-7.
  • 111
    Miquel S, Martín R, Lashermes A, Gillet M, Meleine M, et al. Anti-nociceptive effect of Faecalibacterium prausnitzii in non-inflammatory IBS-like models. Sci Rep. 2016; 6: 19399.
  • 112
    Agostini S, Goubern M, Tondereau V, Salvador-Cartier C, Bezirard V, Lévèque M, et al. A marketed fermented dairy product containing Bifidobacterium lactis CNCM I-2494 suppresses gut hypersensitivity and colonic barrier disruption induced by acute stress in rats. Neurogastroenterol Motil. 2012; 24(4): 376-e172.
  • 113
    Dai C, Guandalini S, Zhao DH, Jiang M. Antinociceptive effect of VSL#3 on visceral hypersensitivity in a rat model of irritable bowel syndrome: a possible action through nitric oxide pathway and enhance barrier function. Mol Cell Biochem. 2012; 362(1-2): 43-53.
  • 114
    Si JM, Yu YC, Fan YJ, Chen SJ. Intestinal microecology and quality of life in irritable bowel syndrome patients. World J Gastroenterol. 2004; 10(12): 1802-5.
  • 115
    Rajilic-Stojanovic M, Biagi E, Heilig HG, Kajander K, Kekkonen RA, Tims S, et al. Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome. Gastroenterology. 2011; 141(5): 1792-801.
  • 116
    Pinn DM, Aroniadis OC, Brandt LJ. Is fecal microbiota transplantation the answer for irritable bowel syndrome? A single-center experience. Am J Gastroenterol. 2014; 109(11): 1831-2.
  • 117
    Johnsen PH, Hilpüsch F, Cavanagh JP, Leikanger IS, Kolstad C, Valle PC, et al. Faecal microbiota transplantation versus placebo for moderate-to-severe irritable bowel syndrome: a double-blind, randomised, placebo-controlled, parallel-group, single-centre trial. Lancet Gastroenterol Hepatol. 2018; 3(1): 17-24.
  • 118
    Cavaletti G, Marmiroli P. Chemotherapy-induced peripheral neurotoxicity. Nat Rev Neurol. 2010; 6(12): 657-66.
  • 119
    Seretny M, Currie GL, Sena ES, Ramnarine S, Grant R, MacLeod MR, et al. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: a systematic review and meta-analysis. Pain. 2014; 155(12): 2461-70.
  • 120
    Zhong S, Zhou Z, Liang Y, Cheng X, Li Y, Teng W, et al. Targeting strategies for chemotherapy-induced peripheral neuropathy: does gut microbiota play a role? Crit Rev Microbiol. 2019; 45(4): 369-93.
  • 121
    Shen S, Lim G, You Z, Ding W, Huang P, Ran C, et al. Gut microbiota is critical for the induction of chemotherapy-induced pain. Nat Neurosci. 2017; 20(9): 1213-6.
  • 122
    Ding W, You Z, Chen Q, Yang L, Doheny J, Zhou X, et al. Gut microbiota influences neuropathic pain through modulating proinflammatory and anti-inflammatory T cells. Anesth Analg. 2021; 132(4): 1146-55.
  • 123
    Yang C, Fang X, Zhan G, Huang N, Li S, Bi J, et al. Key role of gut microbiota in anhedonia-like phenotype in rodents with neuropathic pain. Transl Psychiatry. 2019; 9(1): 57
  • 124
    de Groot PF, Belzer C, Aydin Ö, Levin E, Levels JH, Aalvink S, et al. Distinct fecal and oral microbiota composition in human type 1 diabetes, an observational study. PLoS One. 2017; 12(12): e0188475.
  • 125
    Tang W, Chen X, Liu H, Lv Q, Zou J, Shi Y, et al. Expression of Nrf2 promotes Schwann cell-mediated sciatic nerve recovery in diabetic peripheral neuropathy. Cell Physiol Biochem. 2018; 46(5): 1879-94.
  • 126
    Yan JE, Yuan W, Lou X, Zhu T. Streptozotocin-induced diabetic hyperalgesia in rats is associated with upregulation of Toll-like receptor 4 expression. Neurosci Lett. 2012; 526(1): 54-8.
  • 127
    Liu P, Yuan HB, Zhao S, Liu FF, Jiang YQ, Guo YX, et al. Activation of GABAB receptor suppresses diabetic neuropathic pain through toll-like receptor 4 signaling pathway in the spinal dorsal horn. Mediators Inflamm. 2018; 2018: 6016272.
  • 128
    Thakur V, Gonzalez M, Pennington K, Chattopadhyay M. Viral vector mediated continuous expression of interleukin-10 in DRG alleviates pain in type 1 diabetic animals. Mol Cell Neurosci. 2016; 72: 46-53.
  • 129
    Wagner R, Myers RR. Schwann cells produce tumor necrosis factor alpha: expression in injured and non-injured nerves. Neuroscience. 1996; 73(3): 625-9.
  • 130
    George A, Schmidt C, Weishaupt A, Toyka KV, Sommer C. Serial determination of tumor necrosis factor-alpha content in rat sciatic nerve after chronic constriction injury. Exp Neurol. 1999; 160(1): 124-32.
  • 131
    Shubayev VI, Myers RR. Upregulation and interaction of TNFalpha and gelatinases A and B in painful peripheral nerve injury. Brain Res. 2000; 855(1): 83-9.
  • 132
    Sommer C, Schmidt C, George A. Hyperalgesia in experimental neuropathy is dependent on the TNF receptor 1. Exp Neurol. 1998; 151(1): 138-42.
  • 133
    Iwatsuki K, Arai T, Ota H, Kato S, Natsume T, Kurimoto S, et al. Targeting anti-inflammatory treatment can ameliorate injury-induced neuropathic pain. PLoS One. 2013; 8(2): e57721.
  • 134
    Nadeau S, Filali M, Zhang J, Kerr BJ, Rivest S, Soulet D, et al. Functional recovery after peripheral nerve injury is dependent on the pro-inflammatory cytokines IL-1ß and TNF: implications for neuropathic pain. J Neurosci. 2011; 31(35): 12533-42.
  • 135
    Sorkin LS, Doom CM. Epineurial application of TNF elicits an acute mechanical hyperalgesia in the awake rat. J Peripher Nerv Syst. 2000; 5(2): 96-100.
  • 136
    Wagner R, Myers RR. Endoneurial injection of TNF-alpha produces neuropathic pain behaviors. Neuroreport. 1996; 7(18): 2897-901.
  • 137
    Zoppini G, Faccini G, Muggeo M, Zenari L, Falezza G, Targher G. Elevated plasma levels of soluble receptors of TNF-alpha and their association with smoking and microvascular complications in young adults with type 1 diabetes. J Clin Endocrinol Metab. 2001; 86(8): 3805-8.
  • 138
    González-Clemente JM, Mauricio D, Richart C, Broch M, Caixàs A, Megia A, et al. Diabetic neuropathy is associated with activation of the TNF-alpha system in subjects with type 1 diabetes mellitus. Clin Endocrinol (Oxf). 2005; 63(5): 525-9.
  • 139
    Moriwaki Y, Yamamoto T, Shibutani Y, Aoki E, Tsutsumi Z, Takahashi S, et al. Elevated levels of interleukin-18 and tumor necrosis factor-alpha in serum of patients with type 2 diabetes mellitus: relationship with diabetic nephropathy. Metabolism. 2003; 52(5): 605-8.
  • 140
    Shi X, Chen Y, Nadeem L, Xu G. Beneficial effect of TNF-a inhibition on diabetic peripheral neuropathy. J Neuroinflammation. 2013; 10: 69.
  • 141
    Yamakawa I, Kojima H, Terashima T, Katagi M, Oi J, Urabe H, et al. Inactivation of TNF-a ameliorates diabetic neuropathy in mice. Am J Physiol Endocrinol Metab. 2011; 301(5): E844-52.
  • 142
    Lanza M, Campolo M, Casili G, Filippone A, Paterniti I, Cuzzocrea S, et al. Sodium butyrate exerts neuroprotective effects in spinal cord injury. Mol Neurobiol. 2019; 56(6): 3937-47.
  • 143
    Lanza M, Filippone A, Ardizzone A, Casili G, Paterniti I, Esposito E, et al. SCFA Treatment alleviates pathological signs of migraine and related intestinal alterations in a mouse model of NTG-induced migraine. Cells. 2021; 10(10): 2756.
  • 144
    Vanhoutvin SA, Troost FJ, Kilkens TO, Lindsey PJ, Hamer HM, Jonkers DM, et al. The effects of butyrate enemas on visceral perception in healthy volunteers. Neurogastroenterol Motil. 2009; 21(9): 952-e76.
  • 145
    Du Y, Neng Q, Li Y, Kang Y, Guo L, Huang X, et al. Gastrointestinal autonomic neuropathy exacerbates gut microbiota dysbiosis in adult patients with type 2 diabetes mellitus. Front Cell Infect Microbiol. 2022; 11: 804733.
  • 146
    Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol. 2009; 9(5): 313-23.
  • 147
    Song W, Jiang W, Wang C, Xie J, Liang X, Sun Y, et al. Jinmaitong, a traditional Chinese compound prescription, ameliorates the streptozocin-induced diabetic peripheral neuropathy rats by increasing sciatic nerve IGF-1 and IGF-1R expression. Front Pharmacol. 2019; 10: 255.
  • 148
    Shi Y, Liang XC, Wu QL, Sun LQ, Qu L, Zhao L, et al. Effects of Jinmaitong capsule on ciliary neurotrophic factor in sciatic nerves of diabetes mellitus rats. Chin J Integr Med. 2013; 19(2): 104-11.
  • 149
    Yin DH, Liang XC, Zhao LI, Zhang H, Sun Q, Wang PY, et al. Jinmaitong decreases sciatic nerve DNA oxidative damage and apoptosis in a streptozotocin-induced diabetic rat model. Exp Ther Med. 2015; 10(2): 778-86.
  • 150
    Liang X, Cui L, Guo S. Clinical study on jinmaitong composita on diabetic peripheral neuropathy. Zhongguo Zhong. Xi Yi Jie He Za Zhi. 1999; 19(9): 517-9.
  • 151
    Xie J, Song W, Liang X, Zhang Q, Shi Y, Liu W, et al. Jinmaitong ameliorates diabetic peripheral neuropathy in streptozotocin-induced diabetic rats by modulating gut microbiota and neuregulin 1. Aging (Albany NY). 2020; 12(17): 17436-58.
  • 152
    Xie J, Song W, Liang X, Zhang Q, Shi Y, Liu W, et al. Protective effect of quercetin on streptozotocin-induced diabetic peripheral neuropathy rats through modulating gut microbiota and reactive oxygen species level. Biomed Pharmacother. 2020; 127: 110147.
  • 153
    Cai TT, Ye XL, Yong HJ, Song B, Zheng XL, Cui BT, et al. Fecal microbiota transplantation relieve painful diabetic neuropathy: a case report. Medicine (Baltimore). 2018; 97(50): e13543.

Publication Dates

  • Publication in this collection
    20 Mar 2023
  • Date of issue
    2023

History

  • Received
    26 Aug 2022
  • Accepted
    14 Feb 2023
Instituto Oswaldo Cruz, Ministério da Saúde Av. Brasil, 4365 - Pavilhão Mourisco, Manguinhos, 21040-900 Rio de Janeiro RJ Brazil, Tel.: (55 21) 2562-1222, Fax: (55 21) 2562 1220 - Rio de Janeiro - RJ - Brazil
E-mail: memorias@fiocruz.br