Open access peer-reviewed chapter

Probiotics in Health and Immunity: A First Step toward Understanding the Importance of Microbiota System in Translational Medicine

Written By

Ciro Gargiulo Isacco, Andrea Ballini, Danila De Vito, Angelo Michele Inchingolo, Stefania Cantore, Gregorio Paduanelli, Kieu Cao Diem Nguyen, Alessio Danilo Inchingolo, Gianna Dipalma and Francesco Inchingolo

Submitted: 01 July 2019 Reviewed: 15 July 2019 Published: 14 August 2019

DOI: 10.5772/intechopen.88601

From the Edited Volume

Prebiotics and Probiotics - Potential Benefits in Nutrition and Health

Edited by Elena Franco-Robles and Joel Ramírez-Emiliano

Chapter metrics overview

1,073 Chapter Downloads

View Full Metrics

Abstract

There are mounting evidences showing the relation of chronic inflammatory and autoimmune diseases with the uncontrolled intensification of gut dysbiosis. This position asserts that an elevated presence of pathogens and bacterial, fungal, and viral components is directly involved in inflammatory metabolic diseases with a strong alteration of autoimmune components such as in inflammatory bowel syndrome (IBS), ulcerative colitis, and Crohn’s disease. Furthermore, the increase of unbalanced enteric microbiota is also connected to other types of conditions of metabolic origin such as diabetes mellitus, atherosclerosis, and osteo-degenerative conditions. As a matter of fact, evidence confirmed that gut damages histologically inspected revealed a situation with high expression of pro-inflammatory cytotumor necrosis factor-alpha (TNF-α), and IL1β, IL-2, IL-4, IL-6, and IL-17 together with high level of mucin-2. This chapter focuses on diverse topics related to microbiota dysfunction and systemic health condition and regenerative capacity and the therapeutic role of probiotics in gut health and disease emphasizing the potential beneficial role of probiotics in idiopathic inflammatory metabolic diseases. In brief, outcomes demonstrate that an intimate relationship between microbiota, metabolism, tissue/cellular damages, and regeneration is standing. Within this scenario, the gut certainly plays a big part of the regenerative mechanisms in translational medicine.

Keywords

  • inflammatory bowel syndrome
  • ulcerative colitis
  • Crohn’s disease
  • gut dysbiosis
  • short-chain fatty acids
  • central nervous system
  • experimental autoimmune encephalopathy
  • vagus nerve
  • tumor necrosis factor-alpha
  • IL1β
  • IL-2
  • IL-4
  • IL-6
  • IL-17
  • mucin-2
  • probiotics

1. Introduction

The oral-gastro-intestinal-sex-skin can be classified as unique large and heterogeneous apparatus populated by a huge variety of microorganisms, bacteria, virus, fungi, and other single-celled creatures, that compose the totality of human microbiota that contributes together with bone/skeleton system, to maintain the body energy homeostasis. The human body hosts something like 10–100 trillion microbial cells that coexist in a strict fruitful symbiotic relationship that persists as long as the body is kept in a balanced healthy state [1, 2]. The gut plays an important role in regulating metabolic immune activities. The gut’s essential task is the absorption of nutrients and the synthesis of important micromolecules obtained from food that cannot be assimilated by the stomach and small intestine [1, 2, 3]. Xyloglucans and fructo-oligosaccharides from vegetables and fruits, protein, and lipids; the assimilation of essential vitamins like vitamins B-12, D, and K; and the synthesis of hormones like serotonin from tryptophan amino acid take place right in the gut, thanks to the constant activity of its entire microbiota. The microbiota are able to produce 50–100 mmol·L—1 per day of extremely important short-chain fatty acids (SCFAs), such as acetic, propionic, and butyric acids—and serve as an energy source to the host intestinal epithelium and skeleton [1, 2, 3, 4].

The importance of SCFAs has been well described by several studies during the last decade; the activity of acetic acid, for instance, has been found to be essential against infections, in blood pressure regulation and against sclerotic plaque deposition in arterial walls. The presence of butyric acid is an essential anti-IBS agent due to its immune-modulator properties and anti-inflammatory action, while propionic acid has been found to be important in preventing obesity and diabetes 2 [1, 2, 3, 4].

Although bacteria, viruses, and fungi might be very harmful and dangerous, they are indispensable for life as well. This symbiotic coexistence throughout the millennia made a deep crucial biological impact on human species, and it has become essential not only for survival but for evolution as well. Accumulating evidences have clearly demonstrated how part of these specific microorganisms can resume specific immunomodulatory roles and the way they affect either composition function or migration of various immune cell subpopulations from one site to a different location. For instance, oral macrophages may migrate under the influence of specific signal induction of local microbiota from oral either to the lungs or even the brain passing through the blood brain barrier (BBB) [5, 6, 7, 8, 9, 10].

The outcomes from experiments performed on germ-free (GF) mice confirmed the great role of gut microbiota in the upsurge of immune system deficiencies. GF animals were shown to have compromised Paneth cells and low levels of natural killer (NK) cells, dendritic cells (DCs), and α/β + and γ/δ + T cell populations that play an important role in defense and pathogenesis during inflammation and infection, especially against certain types of malignancies. In addition, GF animals were highly susceptible to frequent infections due to a decline in angiogenin-4 (Ang4), a powerful antimicrobial part of the class of microbicide proteins in Paneth cells [5, 6, 7, 8, 9, 10].

The alteration of the gut microbiota may contribute to open up the invasion of exogenous pathogens that may destabilize the whole intestinal mucosa. The pathogen systematic overgrowth will trigger a cascade of strong inflammatory responses making intestinal mucosa highly susceptible and motile. The chronic inflammation will weaken the endothelial tide junction to the point that the walls become highly relaxed and permeable causing the phenomenon known as “leaky gut” that allows the free, uncontrolled passage of microorganism into the system via the bloodstream and tissues where they start allocating. In fact, the presence of these typical gut residents could be found in eroded, inflamed, and degenerated joints and organs such as the lungs, heart, brain, and liver [11, 12, 13].

Advertisement

2. Gut dysbiosis: a modality to understand neurodegenerative diseases: the disruption of blood-brain barrier (BBB) may explain the gut-oral-brain axis relationship

The high and uncontrolled levels of pathogenic microorganism colonizing the gut contribute to a condition known as dysbiosis [14]. Since few years the dysbiosis has been associated with a variety of degenerative patterns that tend to subvert the metabolic/neuro/hormonal/immune axis contributing to a variety of disorders that round to different body systems ranging from skeleton, cardiovascular, to neuro system. There are several mechanisms proposed that are able to trigger this state of systemic disorders; one of the possibilities is linked to bacterial metabolites and immune-modulating mediators that contribute to the high permeability of intestinal mucosa allowing local pathogens to get through the mucosal barriers triggering a huge variety of immune responses. A second and though related mechanism is the sabotage of SCFAs’ production; the consequences of this mechanism are the abrupt breakdown of energy balance mechanism, a reduction of cell-bacteria signaling pathway, and the worsening of epithelial cell layer integrity due to the decreased production of tight junction proteins which allows the translocation of LPS into the submucosa as well. The significant presence of pro-inflammatory cytokines and interleukins such as TNFα, IFN-γ, IL-1β, IL-2, IL-4, IL-5, and IL-6 is the peculiar trait of a dysbiotic gut (Figure 1) [14, 15, 16]. A third way of dysbiosis transmission is through the vagus nerve (VN), the main component of parasympathetic nervous system (PSN) which also constitutes an effective bridge of the gut/CNS axis. This hypothesis, today supported by a concrete line of evidences, proposes the existence of a reciprocal interference way between the CNS and gut through the VN. In this view the VN is able to perceive microbiota movement, grade of activity, and therefore degradation; on the other hand, pathogens once out from the gut mucosa barrier are able to communicate and move to the CNS through the VN pathway [14, 15, 16, 17, 18].

Figure 1.

The gut microbiota is a very dynamic ecosystem. The entire gut microbiota is composed of different sub-environments with unique features like niches with specific microbes and tissue interactions. The large intestine represents the more populated area and performs the highest variety of biotransformation under the guide of specific gene expression in charge of enzymes necessary for highly specific biotransformation necessary of the SCFAs. The local flora is crucial for the local microbiome homeostasis, and the whole chain of bio-reaction takes place in spaces with a specific mean pH of 6.5–7. The changes in local balance homeostasis and in pH negatively impact on the mucosa shield that repair the outside and inside permeability gradient. Once the stability and the equilibrium between all the components are broken, the gut walls become fragile under the constant attack of local immune cells that start to deteriorate the integrity of both endothelial wall and mucosa shield that induce on medium long term and accumulation of pro-inflammatory endotoxins, bacteria free passage into the system, and low antimicrobial peptide production with a consequent high gastrointestinal motility.

These essential structural alterations are at the base of neurodegenerative pathologies. Though it is a unique pathological aspect, we may see the presence of a common configuration indeed, which is a shared neurological chronic inflammatory pattern. In all these cases, the chronic neuro-inflammatory condition is characterized by an abnormal hyperactive behavior of neural immune cells, the microglia, known as macrophages of the brain [18, 19]. The chronic inflammatory state that from the gut opens up the pathway of pathogenic microbiota invasion all the way through oral and brain compartment, which is the hallmark of neurodegenerative disorders’ dynamic pathogenesis. Patients with Parkinson’s disease (PD), Alzheimer’s (AD), multiple sclerosis (MS), or amyotrophic lateral sclerosis all present a variety of disturbances in intestinal microbial compared to healthy individuals. Neurodegenerative-affected patients’ intestinal and fecal analysis showed a clear clinical picture of microbiota dysbiosis. The test outcomes showed high level of coliform and gram-negative bacteria from Ralstonia genus concomitantly with low critical level of anti-inflammatory strains related to Blautia, Coprococcus, and Roseburia genera. Another indicator was also noted; it was the low presence of Prevotella generally seen as beneficial bacteria, involved in the metabolism of plant polysaccharides and vitamins strictly associated with the production of neuroactive SCFAs, such as GABA [20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31]. The Prevotella spp. is associated with mucin-type O-glycan production which is extremely important in the integrity of gut epithelial barrier; the absence of this mucin type (mucin-2 specifically) tends to compromise the correct homeostatic balance of the local microbiota, increasing intestinal permeability, a clinical feature associated with both microbial dysbiosis and neurodegenerative diseases [32, 33, 34].

Disruption of the BBB is a hallmark in individuals with neurodegenerative diseases that contributes to a steady and progressive death of dopaminergic neurons in the CNS. The BBB is a part of a systemic condition that eventually allows the invasion of pathogens and immune agents from a dysbiotic gut into the CNS. However, damages are also due to a series of changes that weaken the integrity of microvasculature and blood vessels; these modifications are mainly due to nutritional impairment as a consequence of gut microbiota disturbances that cause low-level intake of important nutrients. Deficiencies in vitamins like C, K, D, and folates responsible for low hydroxylation for the formation of chondro-sulfate necessary for healthy microvessel endothelial walls, the augmentation of free radicals, and depletion in oxygen contents and nitrogen, matrix metalloproteinases (MMPs), cyclooxygenases (COXs), and hypoxia-inducible factor-1α (HIF-1α) are all linked with BBB disruption as neuro-inflammatory responses tend to increase and evolve [35].

Thus the scenario existing in the great majority of neurodegenerative pathologies presents a combination of higher permeability of the intestinal barriers and the BBB, inducing a greater access between gut microbiota and the CNS compartment. Experiments conducted with the use of high dose of minocycline antibiotic are well known to have an impact on specific gut and oral invasive bacteria; the post-administration results showed significant protection on LPS-induced PD in mice data confirmed by a significant amelioration of neuro-inflammatory markers such as TNF-α expression, IL-1α expression, and microglia activation and a substantial amelioration of astrocyte loss with an increased number of surviving dopaminergic neurons compared to control LPS only-injected mice [36, 37]. It follows that a correct use of antibiotics generally known to alter gut microbial diversity may disclose a positive immune protective side effect on inflammatory mechanism existent in PD patients [36, 37, 38]. Several other outcomes have shown the beneficial effects of oral antibiotic, minocycline, and tetracycline, in CNS degenerative condition like the experimental autoimmune encephalopathy (EAE) disease and MS. It was found a significant increase of IL-10 expression concomitant with a favorable increase of a subset of invariant NK T cells and in patients with MS, and there was a substantial reduction of CNS deteriorations [38, 39, 40, 41, 42].

2.1 Crosslink between microbiota dysbiosis and osteo-decay

Aging brings generally substantial physiological alterations—hormonal, humoral, and physical—that involves the entire homeostatic organization of the human body. Of course the GI tract and its microbiota as well undergo through profound changes that under the variations of dietary influences bring to a general decline of cognitive and immune activities. With aging, the gut microbiota lost bacterial balanced diversity with an increase of “pathogenic Proteobacteria” vs. a continuing, steady, and progressive lower level of “friend bacteria” such as Firmicutes, Faecalibacterium prausnitzii, and Actinobacteria (mainly bifidobacteria) [42, 43, 44, 45].

Another important feature of gut microbiota is the ability to modulate genes that can be seen either on regulation or variation; this is one of the main factors that may explain the influences that gut microbiota eventually exert on bone development and on bone-related diseases such as osteoporosis, osteopenia, or the different types of arthritis. The delicate homeostatic balance that regulates bone formation and resorption is partly played by the activity of intestinal microorganisms. This activity is basically performed through the interaction with endocrine/nervous system axis; thereby the hormonal activity such as serotonin, cortisol, and sex hormones and several growth factors affect bone mass in mice and humans. In addition, bone marrow stem cells, circulatory stem cells, and stem cells from bone marrow niche are highly sensitive to gut microenvironment condition which eventually affects the differentiation process toward either osteoblasts or osteoclasts. In this case it has been proven that the metabolic pathway compartment which involves the ribosome activity, glycolysis, oxidative phosphorylation, carbon metabolism, and mitochondria ATP are fully responsible of regulating MSCs’ functionality, growth, proliferation, and differentiation [45, 46].

This important connection has also been confirmed by Xiao and colleagues; they were able to highlight through the single-cell RNA-sequencing analysis the existing connection between the gut microbiota, BM-MSCs, and bone metabolic functionality. The presence of several factors such as the HIF-1 together with the expression of infection/inflammatory signaling pathways could be the scattering patterns that influence MSC mobility and immunomodulation. These outcomes showed how HIF-1 signaling is involved in BM-MSC immunomodulation. In fact, the HIF-1 is notoriously known as a triggering factor of inflammatory transcription factor NFκB and an active regulator of specific cytokine and chemokine recruitment in inflammation and infection situations. The chronic presence of an inflammatory response under the triggering activity of pro-inflammatory cytokines such as the TNF superfamily IL-1β, IL-2, IL-4, IL-6, and IL-17 can deeply disturb the osteoclast and osteoblast balance, typically resulting in a net hyper-osteoclast activity and thus bone loss. While there is an evident mechanical effect on the bone where these cytokines stimulate osteoclast differentiation with a consequent upregulation of RANKL expression in progenitor osteoblasts together with a higher RANKL expression, a concomitant nonmechanical effect under the downregulation of specific deficit due to a metabolic inability of vitamin K and vitamin D synthesis in the intestinal lumen should be mentioned (Figure 2) [47, 48, 49, 50, 51, 52, 53, 54].

Figure 2.

The gut dysbiosis is one of the main contributors in osteo-degenerative conditions. The dysbiotic microenvironment increase the viability of systemic pro-inflammatory cytokines and interleukins generating three main anti-regenerative patterns, the increase of pH acidic level, decrease of the differentiating pathway from MSCs and SCs toward osteoblasts, and hyper-expression of osteoclast activity. The dysbiosis generates a defective absorption mechanism of important nutrients for bone homeostasis like vitamins, among them K and D, and hormones such as serotonin, testosterone, and estrogen. The prerogative of this condition is a cascade of events that will involve systemically and progressively the whole vital activity of cells, tissues, and systems of the organism.

2.2 Probiotic efficacy in therapy and clinically use

The potential impact of the therapeutic effect of probiotic on a dysbiotic situation could not be seen without taking in consideration a change of lifestyle. Diet habits, stress, poor healthy conditions, and lack of exercise can significantly impact the gut microbiota stability [55, 56, 57]. There are many evidences that nutritional habits based on “Western diet” composed of huge additive and animal fats, processed glucose and excessive quantities of hypercaloric nutritional facts, low contents of fresh food, and low level of vitamins and minerals, essential for our body, all negatively affect the correct balance of gut microbiota, which eventually lead to the insurgence of metabolic dysfunctions. It is also well known that these bad behaviors are associated with an increased risk of developing several chronic diseases that may attack oral microbiota and vaginal microbiota that recent study findings have indicated as an independent risk factor for severe neurodegenerative conditions [9, 58, 59, 60].

By definition, probiotic refers to large and diverse types of microbes both commensal that normally reside in the gut and exogenous that may migrate through the intestine following food or diet and supplement consumption. Probiotics might be composed of different microbial strains, the most common include species of Lactobacillus, Bifidobacterium, Streptococcus, and yeast Saccharomyces species [61].

As previously mentioned, currently there is a great interest on the use of specific probiotics as therapeutic tool to be associated as clinical approach toward immune system pathologies that may include autoimmune conditions that may attack nerves, bone, and bowel. Given the prevalence of probiotic use, the effects of probiotics on bone health is of significant interest.

Significant positive clinical outcomes have been obtained in numerous studies conducted on CNS inflammatory condition that have therapeutically used different types of probiotic strains. The results showed a reduction of CNS inflammatory level and progression; these outcomes were eventually explained by the capacity of certain strains (Lactobacillus species including bacteria like the Pediococcus acidilactici, Bifidobacterium bifidum, Bifidobacterium animalis, Streptococcus thermophiles, and Bifidobacterium infantis 35,624) to modulate the expression of T-regs, B-regs, and IL-10 production such as [62, 63, 64].

In addition, an experiment with genetically engineered microbial strains such as Lactococcus lactis capable of expressing heat shock protein 65 obtained from another strain like the Mycobacterium leprae was seen highly efficient in reducing EAE symptoms and disease progression [63, 64]. The beneficial outcomes in this study were associated with a decrease in IL-17 pro-inflammatory interleukin with a parallel growth of IL-10 evaluated in the mesenteric lymph node and spleen cell cultures. Furthermore, mice showed a significant higher level of endogenous CD4 + Foxp3+ regulatory T-regs and CD4 + LAP+ (latency-associated peptide). These results might be also sustained by a higher production of SCFAs that, as stated by Opazo and colleagues, were seen to induce either a decrease in RORγt, a biomarker of IL-17, or IL-23 with a higher production of IL-10 and IL-12 with a similar beneficial effect on both EAE and IBS [61, 62, 63, 64, 65, 66].

Therefore beneficial homeostatic-metabolic effect of specific probiotic strains can be seen on different systems such as the cardiovascular, immune, and CNS. For instance, few strains conserve a natural ability of inhibiting the insurgence of hypercholesteremia in both mice and human. In fact the use of Lactobacillaceae strains such as L. acidophilus, Bifidobacterium bifidum, and L. plantarum Lp9 strain showed a significant role in lowering the cholesterol level under in vivo conditions thanks to their ability of secreting functional bile salt hydrolase (BSH), an enzyme crucial in the protection against the insurgence of bad cholesterol in the host. Genomic analysis has indicated that Lactobacillaceae especially L. plantarum contain the highest presence of BSH genes. Intriguingly, milk fermented by L. plantarum NTU 102 revealed to have a high significant efficacy on total cholesterol and LDL cholesterol levels though in presence of individuals undergone a cholesterol-rich diet [67, 68, 69, 70].

Major depressive disorders have been seen even today as a consequence of decreased serotonin level; therefore, the therapeutic strategy has mainly concentrated on producing medication, which focused on serotonin only. The major treatments are based on a class of drugs known as selective serotonin reuptake inhibitors (SSRIs). These SSRIs stimulate the serotonin uptake between neurons, and, though it has been seen some improvements, the medium long-term use has produced serious side effects on gut homeostatic balance with severe metabolic disturbances. Nowadays, as above mentioned, following the fact that current researches have established associations between gut microbes, digestive function, and mental well-being especially under the fact that serotonin is synthesized in the gut by the combined activity of different microbiota strain such as Lactobacillus. The connection was firstly seen in IBS patient who also manifests clear clinical signs of depressive disorders; the analysis of gut microbiota from these patients showed a very low level of Lactobacillus strains versus healthy subjects that might be explained by the increased expression of serotonin transporter (SERT) [71, 72, 73, 74].

Overall the data on this specific topic all have evidenced the positive effects of probiotics in CNS health. These effects are explained by the ability of probiotics to directly interact with fundamental metabolic agents either within the gut or outside that eventually explain the Gut-CNS axis. The use of probiotics and in specific the Lactobacillus strains showed that mice fed with these probiotics revealed a better capacity of reabsorbing tryptophan amino acid the precursor or serotonin, the re-established normal level of those hormones strictly related to stress deviances and depression such as the adrenocorticotropic hormone (ACTH), corticosterone, adrenaline, noradrenaline and the re-increase expression of brain-derived neurotrophic factor (BDNF) a marker that indicate a neuronal health and memory functionality [74, 75, 76, 77, 78, 79].

To conclude, the higher permeability of gut or “leaky gut” intensifies the fee passage of endotoxins such as the LPS and other forms of molecules and pathogens to leak into the bloodstream and thus in the entire system. The upsurge of these endotoxins, pathogens, and waste molecules eventually trigger the activation of a cascade of immune responses through switching on the Toll-like receptor 4 (TLR4) that mediates the recruitment of T and B lymphocytes together with a huge number of pro-inflammatory cytokines, interleukins, and IgA (Figure 3) [80, 81]. The current position therefore considers the use of probiotics as a therapeutic tool that may exert beneficial effects on the CNS by improving the stability homeostasis and integrity of gut microbiota, decreasing systemic inflammation.

Figure 3.

There is a strict connection between CNS and gut system. The connection takes place through the afferent and efferent pathways of vagus nerve that physically connects both CNS and gut. Both CNS and gut may undergo leaky phenomena; in both cases, the barriers of either CNS or gut become extremely permeable under the chronic attack of both pathogens and immune agents overexpressed on the site. This event may eventually explain degenerative condition of both systems including IBS, ulcerative colitis, depression, PD, AD, and MS.

2.2.1 Probiotics in the treatment of skin and oral mucosa dysbiosis

The skin represents another system where an immense variegation of microbiota environment can be found. Skin diseases caused by disturbances at the level of local microbiota that also showed to have strict connection with gut dysbiosis are quite exhaustive in explaining these malevolence patterns. Psoriatic lesions show a very specific histopathological conformation which present highly infiltrated immune cells like the CD3+ T cells and dendritic cells (DCs). Psoriasis showed to have a genetic family trait prevalent in twins; researchers have spotted 36 genetic loci associated with PS susceptibility 1 (PSORS1) locus on chromosome 6p21.3 [82, 83]. Data confirmed that most of them are directly involved in the overexpression of those genes that regulate part of pro-inflammatory innate immune responses such as the NFkB activation and interleukin (IL)-23 signaling pathway. Intriguingly a 2018 study performed on mice proved the use of two specific probiotic Lactobacillus strains, the L. salivarius L305 and L. rhamnosus L307, in alleviating the clinical symptoms of psoriasis through inhibiting the aggressive effect of pro-inflammatory cytokines and interleukins like TNF-α, IL-1β, IL-6, IL-17, and IL-22 and promoting the anti-inflammatory/modulatory activity of IL-4 and IL-10 [84].

In oral dysbiosis, we are facing a similar inflammatory arrangement; oral diseases manifest with high-grade inflammatory patterns that spread from the gums to the adjacent structures gradually destroying the supporting tissues of the teeth, both ligaments and alveolar bones, causing early loss. Similarly to psoriasis in periodontitis, we may encounter multifactorial condition due to a combination of genetic variants triggered by the initial subgingival dysbiosis and then become highly susceptible to wider disease progression [85].

The gram-negative bacteria such as Porphyromonas gingivalis, Tannerella forsythia, and Aggregatibacter actinomycetemcomitans are be able to migrate into the system either down to the heart, lungs, and sex apparatus or are capable to enter into the brain via the bloodstream or via infected periodontal sites. Histopathological analysis has confirmed these bacteria almost everywhere in atheromatous plaques, the vagina, amniotic fluid, rheumatoid arthritis bioptic samples, and brain plaques typical of neurodegenerative diseases AD, PD, and MS [86, 87, 88, 89, 90].

As can be seen from published studies, different strains of probiotics have been used for the treatment of periodontitis. Lactobacillus strains are the commonest used in the majority of high-grade inflammatory disease. The use of L. salivarius in combination with L. rhamnosus and B. subtilis together with L. reuteri and L. brevis probiotics has shown the most promising results. High-positive results were also obtained by Laleman and colleagues in using Streptococcus oralis KJ3, Streptococcus uberis KJ2, and Streptococcus ratti JH145 [91, 92].

Therefore an associated altered gut microbiota may lead to chronic gut dysbiosis and propagation of systemic injuries that involve cells, tissues, system, and the intrinsic dysfunction of the regenerative mechanism.

Advertisement

3. Conclusion

In summary, the present chapter reveals that gut microbiota and a correct use of probiotic may play important pleiotropic functions on several levels and systems. It is now clear that there is a bidirectional interaction between microbiota and nervous system, microbiota and immunity, microbiota and bones, and eventually microbiota and mitochondria. Probiotics are getting more and more attention due to the increase of evidence of their benefits in many degenerative disorders. It shows the capacity of microbiota to restore gut and vaginal and oral microbiota, thus attenuating various severe inflammatory responses. All these findings suggest that probiotics could play a role in clinical procedure and therapy approaches to decrease the risk of morbidity and mortality related to CNS diseases, cardiovascular diseases, and bone degenerations. The shared information presented on this chapter may also demonstrate that the traditional view on gut microbiota and microbiome has changed and may be eventually useful as a prospective medium for the delivery of superior, more precise, and personalized treatments in the achievement of better protective health benefits for a more and more aging society.

Advertisement

Conflicts of interest

The authors declare no conflicts of interest.

Advertisement

Author contribution

The authors contributed equally to this work.

References

  1. 1. Baohong W, Mingfei Y, Longxian L, Zongxin L, Lanjuan L. The human microbiota in health and disease. Engineering. 2017;3(1):71-82
  2. 2. Moya A, Ferrer M. Functional redundancy-induced stability of gut microbiota subjected to disturbance. Trends in Microbiology. 2016;24(5):402-413
  3. 3. Dumas ME, Barton RH, Toye A, Cloarec O, Blancher C, Rothwell A. Metabolic profiling reveals a contribution of gut microbiota to fatty liver phenotype in insulin-resistant mice. Proceedings of the National Academy of Sciences of the United States of America. 2006;103:12511-12516
  4. 4. Borycka-Kiciak K, Banasiewicz T, Rydzewska G. Butyric acid – A well-known molecule revisited. Przegla̜d Gastroenterologiczny. 2017;12(2):83-89
  5. 5. Al-Lahham SH, Peppelenbosch MP, Roelofsen H, Vonk RJ, Venema K. Biological effects of propionic acid in humans; metabolism, potential applications and underlying mechanisms. Biochimica et Biophysica Acta. 2010;1801(11):1175-1183
  6. 6. Acheson DW, Luccioli S. Microbial-gut interactions in health and disease. Mucosal immune responses. Best Practice & Research. Clinical Gastroenterology. 2004;18(2):387-404
  7. 7. Thomas S, Izard J, Walsh E, Batich K, Chongsathidkiet P, Clarke G, et al. The host microbiome regulates and maintains human health: A primer and perspective for non-microbiologists. Cancer Research. 2017:1-31
  8. 8. Round L, Mazmanian SK. The gut microbiome shapes intestinal immune responses during health and disease. Nature Reviews Immunology. 2009;9(5):313-323
  9. 9. Holly MK, Smith JG. Paneth cells during viral infection and pathogenesis. Viruses. 2018;10(5):225
  10. 10. Ballini B, Santacroce L, Cantore S, Bottalico L, Dipalma G, Topi S, et al. Probiotics efficacy on oxidative stress values in inflammatory bowel disease: A randomized double-blinded placebo-controlled pilot study. Endocrine, Metabolic & Immune Disorders Drug Targets. 2019;19(12):1-12
  11. 11. Lannes N, Eppler E, Etemad S, Yotovski P, Filgueira L. Microglia at center stage: A comprehensive review about the versatile and unique residential macrophages of the central nervous system. Oncotarget. 2017;8(69):114393-114413
  12. 12. Sonnenburg JL, Bäckhed F. Diet-microbiota interactions as moderators of human metabolism. Nature. 2016;535:56-64
  13. 13. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nature Reviews. Immunology. 2009;9:313-323
  14. 14. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57:1470-1481
  15. 15. Spielman LJ, Gibson DL, Klegeris A. Unhealthy gut, unhealthy brain: The role of the intestinal microbiota in neurodegenerative diseases. Neurochemistry International. 2019;120:149-163
  16. 16. Barrett E, Ross RP, O’Toole TW, Fitzgerald GF, Stanton C. Gamma-Aminobutyric acid production by culturable bacteria from the human intestine. Journal of Applied Microbiology. 2005;113:411-417
  17. 17. Bergstrom KS, Xia L. Mucin-type O-glycans and their roles in intestinal homeostasis. Glycobiology. 2013;23:1026-1037
  18. 18. Bonaz B, Sinniger V, Pellissier S. Anti-inflammatory properties of the vagus nerve: Potential therapeutic implications of vagus nerve stimulation. The Journal of Physiology. 2016;594:5781-5790
  19. 19. Russo R, Cristiano C, Avagliano C, De Caro C, La Rana G, Raso GM, et al. Gut-brain axis: Role of lipids in the regulation of inflammation, pain and CNS diseases. Current Medicinal Chemistry. 2018;25(32):3930-3952
  20. 20. Block ML, Hong JS. Microglia and inflammation-mediated neurodegeneration: Multiple triggers with a common mechanism. Progress in Neurobiology. 2005;76:77-98
  21. 21. Zhang YG, Wu S, Yi J, Xia Y, Jin D, Zhou J, et al. Target intestinal microbiota to alleviate disease progression in amyotrophic lateral sclerosis. Clinical Therapeutics. 2017;39:322-336
  22. 22. Zhang R, Miller RG, Gascon R, Champion S, Katz J, Lancero M, et al. Circulating endotoxin and systemic immune activation in sporadic amyotrophic lateral sclerosis (sALS). Journal of Neuroimmunology. 2008;206(1-2):121-124
  23. 23. Xu R, Wang Q. Towards understanding brain-gut-microbiome connections in Alzheimer’s disease. BMC Systems Biology. 2016;10(3):63
  24. 24. Wu S, Yi J, Zhang YG, Zhou J, Sun J. Leaky intestine and impaired microbiome in an amyotrophic lateral sclerosis mouse model. Physics Reports. 2015;3(4):e12356
  25. 25. Wang D, Ho L, Faith J, Ono K, Janle EM, Lachcik PJ, et al. Role of intestinal microbiota in the generation of polyphenol-derived phenolic acid mediated attenuation of Alzheimer’s disease beta-amyloid oligomerization. Molecular Nutrition & Food Research. 2015;59:1025-1040
  26. 26. Villumsen M, Aznar S, Pakkenberg B, Jess T, Brudek T. Inflammatory bowel disease increases the risk of Parkinson’s disease: A Danish Nationwide cohort study, 1977-2014. Gut. 2018
  27. 27. Villarán RF, Espinosa-Oliva AM, Sarmiento M, De Pablos RM, Argüelles S, Delgado-Cortés MJ, et al. Ulcerative colitis exacerbates lipopolysaccharide-induced damage to the nigral dopaminergic system: Potential risk factor in Parkinson’s disease. Journal of Neurochemistry. 2010;114(6):1687-1700
  28. 28. Toepfer M, Folwaczny C, Klauser A, Riepl RL, Müller-Felber W, Pongratz D. Gastrointestinal dysfunction in amyotrophic lateral sclerosis. Amyotrophic Lateral Sclerosis. 1999;(1):1, 15-19
  29. 29. Rowin J, Xia Y, Jung B, Sun J. Gut inflammation and dysbiosis in human motor neuron disease. Physiological Reports. 2017;5(18):e13443
  30. 30. Holmqvist S, Chutna O, Bousset L, Aldrin-Kirk P, Li W, Björklund T, et al. Direct evidence of Parkinson pathology spread from the gastrointestinal tract to the brain in rats. Acta Neuropathologica. 2014;128(6):805-820
  31. 31. Cersosimo MG, Raina GB, Pecci C, Pellene A, Calandra CR, Gutiérrez C, et al. Gastrointestinal manifestations in Parkinson’s disease: Prevalence and occurrence before motor symptoms. Journal of Neurology. 2013;260(5):1332-1338
  32. 32. Barrett E, Ross RP, O’Toole PW, Fitzgerald GF, Stanton C. γ-Aminobutyric acid production by culturable bacteria from the human intestine. Journal of Applied Microbiology. 2012;113(2):411-417
  33. 33. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504:451-455
  34. 34. Tailford LE, Crost EH, Kavanaugh D, Juge N. Mucin glycan foraging in the human gut microbiome. Frontiers in Genetics. 2015;6:81
  35. 35. Rausch P, Rehman A, Kunzel S, Hasler R, Ott SJ, Schreiber S. Colonic mucosa-associated microbiota is influenced by an interaction of Crohn disease and FUT2 (secretor) genotype. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:19030-19035
  36. 36. Rosenberg GA. Neurological diseases in relation to the blood-brain barrier. Journal of Cerebral Blood Flow and Metabolism. 2012;32(7):1139-1151
  37. 37. Zaura E, Brandt BW, Teixeira de Mattos MJ, Buijs MJ, Caspers MP, Rashid MU, et al. Same exposure but two radically different responses to antibiotics: Resilience of the salivary microbiome versus long-term microbial shifts in feces. MBio. 2015;6:e01693-e01615
  38. 38. Tomas-Camardiel M, Rite I, Herrera AJ, de Pablos RM, Cano J, Machado A, et al. Minocycline reduces the lipopolysaccharide-induced inflammatory reaction, peroxynitrite-mediated nitration of proteins, disruption of the blood-brain barrier, and damage in the nigral dopaminergic system. Neurobiology of Disease. 2004;16:190-201
  39. 39. Sampson TR, Debelius JW, Thron T, Janssen S, Shastri GG, Ilhan ZE, et al. Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease. Cell. 2016;167:1469-1480
  40. 40. Yokote H, Miyake S, Croxford JL, Oki S, Mizusawa H, Yamamura T. NKT cell-dependent amelioration of a mouse model of multiple sclerosis by altering gut flora. The American Journal of Pathology. 2008;173:1714-1723
  41. 41. Popovic N, Schubart A, Goetz BD, Zhang SC, Linington C, Duncan ID. Inhibition of autoimmune encephalomyelitis by a tetracycline. Annals of Neurology. 2002;51:215-223
  42. 42. Colpitts SL, Kasper LH. Influence of the gut microbiome on autoimmunity in the central nervous system. Journal of Immunology. 2017;198(2):596-604
  43. 43. Odamaki T, Kato K, Sugahara H, Hashikura N, Takahashi S, Xiao JZ, et al. Age-related changes in gut microbiota composition from newborn to centenarian: A cross-sectional study. BMC Microbiology. 2016;16:90
  44. 44. Salazar N, López P, Valdés L, Margolles A, Suárez A, Patterson AM, et al. Microbial targets for the development of functional foods accordingly with nutritional and immune parameters altered in the elderly. Journal of the American College of Nutrition. 2013;32:399-406
  45. 45. Ticinesi A, Tana C, Nouvenne A, Prati B, Lauretani F, Meschi T. Gut microbiota, cognitive frailty and dementia in older individuals: A systematic review. Clinical Interventions in Aging. 2018;13:1497-1511
  46. 46. Salazar N, Valdés-Varela L, González S, Gueimonde M, de Los Reyes-Gavilán CG. Nutrition and the gut microbiome in the elderly. Gut Microbes. 2017;8:82-97
  47. 47. Xiao E, He L, Wu Q , et al. Microbiota regulates bone marrow mesenchymal stem cell lineage differentiation and immunomodulation. Stem Cell Research & Therapy. 2017;8(1):213
  48. 48. Nicholson JK, Holmes E, Kinross J, Burcelin R, Gibson G, Jia W, et al. Host–gut microbiota metabolic interactions. Science. 2012;336(6086):1262-1267
  49. 49. Walmsley SR, Print C, Farahi N, Peyssonnaux C, Johnson RS, Cramer T, et al. Hypoxia-induced neutrophil survival is mediated by HIF-1alpha-dependent NF-kappaB activity. The Journal of Experimental Medicine. 2005;201(1):105-115
  50. 50. Scortegagna M, Cataisson C, Martin RJ, Hicklin DJ, Schreiber RD, Yuspa SH, et al. HIF-1alpha regulates epithelial inflammation by cell autonomous NFkappaB activation and paracrine stromal remodeling. Blood. 2008;111(7):3343-3354
  51. 51. Spees JL, Lee RH, Gregory CA. Mechanisms of mesenchymal stem/stromal cell function. Stem Cell Research & Therapy. 2016;7(1):125
  52. 52. Szychlinska MA, Di Rosa M, Castorina A, Mobasheri A, Musumeci G. A correlation between intestinal microbiota dysbiosis and osteoarthritis. Heliyon. 2019;5(1):e01134
  53. 53. Lam J et al. TNF-alpha induces osteoclastogenesis by direct stimulation of macrophages exposed to permissive levels of RANK ligand. The Journal of Clinical Investigation. 2000;106(12):1481-1488
  54. 54. Walsh NC, Gravallese EM. Bone remodeling in rheumatic disease: A question of balance. Immunological Reviews. 2010;233(1):301-312
  55. 55. Romas E et al. Expression of osteoclast differentiation factor at sites of bone erosion in collagen-induced arthritis. Arthritis and Rheumatism. 2000;43(4):821-826
  56. 56. Huxley RR, Ansary-Moghaddam A, Clifton P, Czernichow S, Parr CL, Woodward M. The impact of dietary and lifestyle risk factors on risk of colorectal cancer: A quantitative overview of the epidemiological evidence. International Journal of Cancer. 2009;125:171-180
  57. 57. Rothschild D, Weissbrod O, Barkan E. Environment dominates over host genetics in shaping human gut microbiota. Nature. 2018;555:210-215
  58. 58. Warburton DE, Nicol CW, Bredin SS. Health benefits of physical activity: The evidence. CMAJ. 2006;174:801-809
  59. 59. Inchingolo F, Dipalma G, Cirulli N, Cantore S, Saini RS, Altini V, et al. Microbiological results of improvement in periodontal condition by administration of oral probiotics. Journal of Biological Regulators and Homeostatic Agents. 2018;32(5):1323-1328
  60. 60. Cantore S, Ballini A, De Vito D, Abbinante A, Altini V, Dipalma G, et al. Clinical results of improvement in periodontal condition by administration of oral probiotics. Journal of Biological Regulators and Homeostatic Agents. 2018;32(5):1329-1334
  61. 61. Kim D, Yoo SA, Kim WU. Gut microbiota in autoimmunity: Potential for clinical applications. Archives of Pharmacal Research. 2016;39:1565-1576
  62. 62. Opazo MC, Ortega-Rocha EM, Coronado-Arrázola I, et al. Intestinal microbiota influences non-intestinal related autoimmune diseases. Frontiers in Microbiology. 2018;9:432
  63. 63. Kwon HK, Kim GC, Kim Y, Hwang W, Jash A, Sahoo A, et al. Amelioration of experimental autoimmune encephalomyelitis by probiotic mixture is mediated by a shift in T helper cell immune response. Clinical Immunology. 2013;146(3):217-227
  64. 64. Rezende RM, Oliveira RP, Medeiros SR, Gomes-Santos AC, Alves AC, Loli FG, et al. Hsp65-producing Lactococcus lactis prevents experimental autoimmune encephalomyelitis in mice by inducing CD4+LAP+ regulatory T cells. Journal of Autoimmunity. 2013;40:45-57
  65. 65. Mizuno M, Noto D, Kaga N, Chiba A, Miyake S. The dual role of short fatty acid chains in the pathogenesis of autoimmune disease models. PLoS One. 2017;12(2):e0173032
  66. 66. Takata KT, Tomita T, Okuno M, Kinoshita T, Koda JA, Honorat M, et al. Dietary yeasts reduce inflammation in central nerve system via microflora. Annals of Clinical Translational Neurology. 2015;2:56-66
  67. 67. Konieczna P, Groeger D, Ziegler M, Frei R, Ferstl R, Shanahan F, et al. Bifidobacterium infantis 35624 administration induces Foxp3 T regulatory cells in human peripheral blood: Potential role for myeloid and plasmacytoid dendritic cells. Gut. 2012;61:354-366
  68. 68. Mohania D, Kansal VK, Shah D, Nagpal R, Kumar M, Gautam SK, et al. Therapeutic effect of probiotic dahi on plasma, aortic, and hepatic lipid profile of hypercholesterolemic rats. Journal of Cardiovascular Pharmacology and Therapeutics. 2013;18(5):1-8
  69. 69. Brandvold KR, Weaver JM, Whidbey C, Wright AT. A continuous fluorescence assay for simple quantification of bile salt hydrolase activity in the gut microbiome. Scientific Reports. 2019;9(1359):1-7
  70. 70. Liang L, Yi Y, Lv Y, Qian J, Lei X, Zhang G. A comprehensive genome survey provides novel insights into bile salt hydrolase (BSH) in Lactobacillaceae. Molecules. 2018;23(5):1157
  71. 71. Guo Z, Liu XM, Zhang QX, Shen Z, Tian FW, Zhang H, et al. Influence of consumption of probiotics on the plasma lipid profile: A meta-analysis of randomised controlled trials. Nutrition, Metabolism, and Cardiovascular Diseases. 2011;21:844-850
  72. 72. Cao YN, Feng LJ, Liu YY, et al. Effect of Lactobacillus rhamnosus GG supernatant on serotonin transporter expression in rats with post-infectious irritable bowel syndrome. World Journal of Gastroenterology. 2018;24(3):338-350
  73. 73. Zhang ZF, Duan ZJ, Wang LX, Yang D, Zhao G, Zhang L. The serotonin transporter gene polymorphism (5-HTTLPR) and irritable bowel syndrome: A meta-analysis of 25 studies. BMC Gastroenterology. 2014;14:23
  74. 74. Wheatcroft J, Wakelin D, Smith A, Mahoney CR, Mawe G, Spiller R. Enterochromaffin cell hyperplasia and decreased serotonin transporter in a mouse model of postinfectious bowel dysfunction. Neurogastroenterology and Motility. 2005;17:863-870
  75. 75. Wallace CJK, Milev R. The effects of probiotics on depressive symptoms in humans: A systematic review [published correction appears in annals of general psychiatry 2017 Mar 7;16:18]. Annals of General Psychiatry. 2017;16:14
  76. 76. Ait-Belgnaoui A, Durand H, Cartier C, et al. Prevention of gut leakiness by a probiotic treatment leads to attenuated HPA response to an acute psychological stress in rats. Psychoneuroendocrinology. 2012;37(11):1885-1895
  77. 77. Ait-Belgnaoui A, Colom A, Braniste V. Probiotic gut effect prevents the chronic psychological stress-induced brain activity abnormality in mice. Neurogastroenterology and Motility. 2014;26(4):510-520
  78. 78. Bravo JA, Forsythe P, Chew MV, et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. PNAS. 2011;108(38):16050-16055
  79. 79. Stetler C, Miller GE. Depression and hypothalamic–pituitary–adrenal activation: A quantitative summary of four decades of research. Psychosomatic Medicine. 2011;73(2):114-126
  80. 80. Sen S, Duman R, Sanacora G. Serum brain-derived neurotrophic factor, depression, and antidepressant medications: Meta-analyses and implications. Biological Psychiatry. 2008;64(6):527-532
  81. 81. Soares JB, Pimentel-Nunes P, Roncon-Albuquerque R, Leite-Moreira A. The role of lipopolysaccharide/toll-like receptor 4 signaling in chronic liver diseases. Hepatology International. 2010;4(4):659-672
  82. 82. Chamian F, Lowes MA, Lin SL, Lee E, Kikuchi T, Gilleaudeau P, et al. Alefacept reduces infiltrating T cells, activated dendritic cells, and inflammatory genes in psoriasis vulgaris. Proceedings of the National Academy of Sciences of the United States of America. 2005;102:2075-2080
  83. 83. Lowes MA, Chamian F, Abello MV, Fuentes-Duculan J, Lin SL, Nussbaum R, et al. Increase in TNF-alpha and inducible nitric oxide synthase-expressing dendritic cells in psoriasis and reduction with efalizumab (anti-CD11a). Proceedings of the National Academy of Sciences of the United States of America. 2005;102:19057-19062
  84. 84. Holowacz S, Blondeau C, Guinobert I, Guilbot A, Hidalgo S, Bisson JF. Lactobacillus salivarius L-307 and Lactobacillus rhamnosus LA-305 attenuate skin inflammation in mice. Beneficial Microbes. 2018;9(2):299-309
  85. 85. Armitage GC. Development of a classification system for periodontal diseases and conditions. Annals of Periodontology. 1999;4:1-6
  86. 86. Page RC, Offenbacher S, Schroeder HE, Seymour GJ, Kornman KS. Advances in the pathogenesis of periodontitis: Summary of developments, clinical implications and future directions. Periodontology. 2000;14:216-248
  87. 87. Leon R, Silva N, Ovalle A, Chaparro A, Ahumada A, Gajardo M, et al. Detection of porphyromonas gingivalis in the amniotic fluid in pregnant women with a diagnosis of threatened premature labor. Journal of Periodontology. 2007;78:1249-1255
  88. 88. Forner L, Larsen T, Kilian M, Holmstrup P. Incidence of bacteremia after chewing, tooth brushing and scaling in individuals with periodontal inflammation. Journal of Clinical Periodontology. 2006;33:401-407
  89. 89. Quirke AM, Lugli EB, Wegner N, Hamilton BC, Charles P, Chowdhury M, et al. Heightened immune response to autocitrullinated Porphyromonas gingivalis peptidylarginine deiminase: A potential mechanism for breaching immunologic tolerance in rheumatoid arthritis. Annals of the Rheumatic Diseases. 2014;73:263-269
  90. 90. Stein PS, Steffen MJ, Smith C, Jicha G, Ebersole JL, Abner E, et al. Serum antibodies to periodontal pathogens are a risk factor for Alzheimer’s disease. Alzheimers Dement. 2012;8:196-203
  91. 91. Jayaram P, Chatterjee A, Raghunathan V. Probiotics in the treatment of periodontal disease: A systematic review. Journal of Indian Society of Periodontology. 2016;20(5):488-495
  92. 92. Laleman I, Yilmaz E, Ozcelik O, Haytac C, Pauwels M, Herrero ER, et al. The effect of a streptococci containing probiotic in periodontal therapy: A randomized controlled trial. Journal of Clinical Periodontology. 2015;42(11):1032-1041

Written By

Ciro Gargiulo Isacco, Andrea Ballini, Danila De Vito, Angelo Michele Inchingolo, Stefania Cantore, Gregorio Paduanelli, Kieu Cao Diem Nguyen, Alessio Danilo Inchingolo, Gianna Dipalma and Francesco Inchingolo

Submitted: 01 July 2019 Reviewed: 15 July 2019 Published: 14 August 2019