Next Article in Journal
Changes in Ground Reaction Forces, Joint Mechanics, and Stiffness during Treadmill Running to Fatigue
Next Article in Special Issue
Early Intervention Using Fecal Microbiota Transplantation Combined with Probiotics Influence the Growth Performance, Diarrhea, and Intestinal Barrier Function of Piglets
Previous Article in Journal
Impact of Nonlinear Chemical Reaction and Melting Heat Transfer on an MHD Nanofluid Flow over a Thin Needle in Porous Media
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Gut Microbiota Imbalance is Related to Sporadic Colorectal Neoplasms. A Pilot Study

1
Polypheno Academic Spin Off, University of Bari “A. Moro”, Policlinico, Piazza G. Cesare 11, 70124 Bari, Italy
2
Gastroenterology Unit, Department of Emergency and Organ Transplantation (DETO), University of Bari “A. Moro”, Policlinico, Piazza G. Cesare 11, 70124 Bari, Italy
3
Interdisciplinary Department of Medicine (DIM), University of Bari “Aldo Moro”, Policlinico, Piazza G. Cesare 11, 70124 Bari, Italy
4
Department of Biosciences, Biotechnologies and Biopharmaceuticals, University of Bari “Aldo Moro”, Via E. Orabona 4, 70124 Bari, Italy
5
Experimental Oncology, Scientific Institute for Cancer Care and Research IRCCS “G. Paolo II”, Viale Orazio Flacco, 65, 70124 Bari, Italy
6
School of Technical Medical Sciences, University of Elbasan “A. Xhuvani”, 3001 Elbasan, Albania
7
Ionian Department, Microbiology & Virology Lab, University of Bari “Aldo Moro”, 70124 Bari, Italy
*
Author to whom correspondence should be addressed.
Appl. Sci. 2019, 9(24), 5491; https://doi.org/10.3390/app9245491
Submission received: 21 November 2019 / Revised: 8 December 2019 / Accepted: 11 December 2019 / Published: 13 December 2019
(This article belongs to the Special Issue New Trends in Biosciences)

Abstract

:
(1) Background: Colorectal cancer (CRC) development is sustained by multiple factors including the gut microbiota, as suggested by a growing body of evidence. Most CRCs have a sporadic (non-hereditary) onset and develop from sporadic colorectal adenomas/polyp (SCA/P). In the present study, we investigated the characteristic of anaerobic microorganisms in stool samples obtained from 20 patients with SCA/P and 20 subjects without evidence of proliferative lesions at colonoscopy (Controls). (2) Material and Methods: We designed this clinical trial using adaptive randomization by minimization. Selective culture media and Matrix Assisted Laser Desorption Ionization Time of Flight (MALDI-TOF) mass spectrometry techniques were used to identify the components of microbiota. The data obtained revealed a different variability of gut microbiota in stool samples of controls and SCA/P subjects. (3) Results: The most interesting difference was observed for Bacteroides species, which represent the 50% of all bacterial species identified in the stool samples: two species, Bacteroides stercoris and Parabacteroides distasonis, were found only in the feces from control group, whereas Bacteroides fragilis and Prevotella melaningenica species were presents only in SCA/P patients. Among Gram+ bacteria also, specific species were found in the two groups of feces: Clostridium clostridioforme, Propionibacterium avidum and Pediococcus pentasaceus were identified only in controls, while Eubacterium limosum, Clostridium innocuum and Corybebacterium xerosus were identified in SCA/P stool samples only. (4) Conclusions: Our findings suggest that, compared to control stool samples, a different intestinal microbiota is present in SCA/P stool samples, that may create a micro-environment predisposing for the development of proliferative phenomena. As a consequence, gut microbiota manipulation could be a future target for personalized treatments.

1. Introduction

Colorectal cancer (CRC) represents the third most common cause of cancer mortality worldwide. Experimental evidence supports the role of gut microbiota as an important contributor in the carcinogenic process [1]. The idea concerning the role of microbes in carcinogenesis is not new, but it received compelling evidence over the last two decades, mainly thanks to the studies on Helicobacter pylori [2]. Human colon contains the largest population of bacteria in the body (in excess of 1011 organisms per gram of wet weight) and the majority of these organisms are anaerobes. Among this population, 25% are Bacteroides, bile-resistant, gram negative bacilli [3]. Statistically, the relative abundance of five genera, Bacteroides, Roseburia, Alistipes, Eubacterium and Parasutterella has been identified in human gut microbiota [4]. A healthy balance between host and microorganism is kept for the optimal performance of metabolic and immune functions and to prevent disease development in intestinal and extra-intestinal regions, including the central nervous system [5]. Indeed, gut microbiota plays a pivotal immune function against pathogenic bacteria colonization and invasion and contributes to the maintenance of the intestinal epithelium integrity [6].
Microbiota variations could be principally due to host-related factors including body mass index (BMI), exercise frequency, ethnicity, cultural and dietary habits. As far as the last aspect, recent studies have shown that diet has an important role in shaping the structure of gut microbiota [7,8,9], which anaerobically ferments dietary components that are not completely digested and absorbed in the upper gastrointestinal tract [10]. Metabolites and antigens produced by gut microbiota may have significant roles in influencing the risk of developing colon cancer through their interactions with host metabolism and immunity [11,12].
It is widely accepted that there is no single causative agent identified in CRC [13]. However, there is evidence that a reduction of “protective” bacteria and an increase in some bacteria such as Fusobacterium sp. or Bacteroides/Prevotella, as well as age-related changes in microbiota have an impact on adenoma and cancer development [13]. Wang et al. [4] reported structural fecal bacterial segregations between CRC patients and healthy volunteers. Shen et al. demonstrated a higher abundance of Proteobacteria and a lower abundance of Bacteroidetes in CRC cases compared to healthy controls [14].
Most colorectal carcinomas develop from pre-cancerous lesions, namely sporadic colorectal adenoma/polyp (SCA/P), considered to have malignant potential [15]. At our knowledge, this is the first study comparing the gut microbiota anaerobic components in stool specimens from patients with SCA/P and subjects without precancerous lesions (controls).

2. Material and Methods

2.1. Study Design

The study is aimed to evaluate possible differences in gut microbiota composition comparing patients with pre-cancerous lesions end/or sporadic colorectal adenomas in respect of healty subjects. A clinical trial using adaptive randomization by minimization was been conducted. The protocol study was approved by the Ethics Committee of the University of Bari, Protocol Record Policlinico Hospital, Bari, ClinicalTrials.gov Identifier: NCT03417258.

2.2. Patient Enrollment

We performed an analysis of anaerobic gut microbiota in subjects that performed colonoscopy at the Gastroenterology Unit, Policlinico University Hospital, Bari, and showed pre-cancerous lesions end/or sporadic colorectal adenomas (Group SCA/P, 20 subjects). In addition, the same analysis was performed in 20 subjects, without proliferative lesions, matched for age and sex (controls). All the participants underwent colonoscopy within the previous 1–3 months before the evaluation of their faecal specimens. Written informed consent was obtained from all study subjects.
The inclusion criteria were: subjects of both sexes aged between 50 and 75 years, undergoing colonoscopy. The exclusion criteria were: age < 50 and >75 years, previous diagnosis of colon cancer or inflammatory bowel diseases (IBD), hereditary intestinal tumours, ongoing infections, intake of drugs that could alter the intestinal microbiota during the last 4 weeks, creatinine clearance below 60 mL/min, liver failure.

2.3. Microbiological Study

Fecal samples were collected from each subject and immediately analyzed for the presence of the anaerobic bacteria. The stool samples were inoculated on both Schaedler blood agar and on Bile Esculin Agar (BEA) as selective isolation medium for anaerobic bacteria Bacteroides spp. After incubation at 37 °C for 72 h in an anaerobic jar, strict anaerobes were chosen. Thereafter, after grown on both culture plates, identification of the different colonies was performed by Matrix Assisted Laser Desorption Ionization Time of Flight Mass Spectrometry (MALDI-TOF MS), an innovative technology for the rapid and accurate identification of bacterial and fungal isolates in clinical settings [16]. MALDI-TOF technology has been developed to profile, within minutes instead of the usually required 36–48 h of the traditional approaches, based on the pure culture of the microorganism, bacterial proteins from whole cell extracts and to obtain a bacterial fingerprint able to discriminate microorganisms from different genera and species [17]. The technology is automated, high throughput, and applicable to a broad range of common as well as rare bacteria and fungi.

2.4. Statistical Analysis

Where indicated, individual comparisons were performed using Student’s t-test. The results obtained are expressed as mean ± S.D. Statistical comparison among groups was determined using analysis of variance. Statistical significance was ascribed to the data when p < 0.05.

3. Results

In Table 1, demographical and anthropometric parameters of the patients involved in the study, matched for sex, age and BMI, are reported. No statistical difference was detected between the two reported groups of subjects.
Table 2 describes the different anaerobic species identified in the stool samples of the 40 subjects participating to the study. Our results show that a total of 9 Gram− and 9 Gram+ species of anaerobic bacteria were present in the feces of these subjects. It is worth noting that all species of Gram− anaerobic bacteria are part of the Bacteroides family which represents the 50% (9/18) of all bacterial species identified.
Figure 1 reports the different gut microbiota species (Gram− and Gram+) recognized in the fecal samples of the two groups of subjects. In controls and SCA/P patients the total number of bacterial species identified was similar (7 Gram− and 6 Gram+). However, among Gram- bacteria, all anaerobic bacteria belonging to the Bacteroides genus, two species, Bacteroides stercoris and Parabacteroides distasonis, were found only in the feces from control group, whereas Bacteroides fragilis and Prevotella melaninigenica species were present only in SCA/P patients. Moreover, it appears evident, among the 5 bacterial species present in both groups of subjects, that the detection of Bacteroides ovatus (B.o.), Bacteroides uniformis (B.u.) and Bacteroides vulgatus (B.v.) was tripled, doubled and slightly higher, respectively, in ontrol group as compared to SCA/P patients, while the presence of Parabacteroides merdae (Pa.m.) was double in the SCA/P group as compared to control subjects. Similar presence was evidenced for Bacteroides thetaiotaomicron (B.t.) in both groups of fecal samples.
As far as the Gram+ bacteria identified in the two group of subjects (Figure 1), similarly to what observed in Gram− bacteria, specific bacterial species were found in the two groups: Clostridium clostridioforme (C.c.), Propionibacterium avidum (Pr.a.) and Pedioccus pentasaceus (Pe.p) were identified in control group only, while Eubacterium limosum (Eu.l.), Clostridium innocuum (C.i.) and Corynebacterium xerosus (Cor.x.) were identified in SCA/P feces only.
Among the bacteria presents in both groups of feces, Bifidobacterium spp (Bif.) resulted tripled in control group compared to SCA/P group, while similar was the presence of Clostridium ramosum (C.r.) and Clostridium sardelli (C.s.) in both group of fecal samples.

4. Discussion

Microbiota manipulations by administration of probiotic have been demonstrated to be effective in a number of clinical conditions (e.g., respiratory, oral, urogenital, and intestinal) to ameliorate the history of either infective or non-infective diseases, probably modulating the local and systemic immune functions [18,19,20,21,22,23]. Herein, for the first time, we report a description of the anaerobic population of gut microbiota in stool samples from controls subjects and patients with sporadic colorectal adenoma/polyp (SCA/P). Our attention to the anaerobic species has been driven by numerous data present in literature reporting a strong correlation between stool anaerobic bacteria and CRC. Zhu et al., reviewing the relationship between microbiota and the development of CRC, reported highest levels of carcinogen production in association with gut anaerobic bacteria [23]. Kwong and coll reported that genera Bacteroides, the 25% of anaerobic bacteria [3], are strongly associated with CRC, stating also that CRC development is associated with Bacteroides fragilis [24]. Farther, epidemiological studies reported that incidence and mortality of CRC higher in African American (AA) than in Caucasian American (CA) associated to the more abundant presence of Bacteroidetes in AAs than in CAs patients [25].
Our data revealed that a different gut microbiota pattern, particularly evident for the Bacteroides genus, was found in the two group of subjects studied. However, our data suggest other possible important implications. First of all, the two groups of stools are characterized by different species of bacteria, Bacteroides stercoris and Parabacteroides distasonis for control group and Bacteroides fragilis and Prevotella melaninigenica for SCA/P group. Data from literature assign specific roles to these bacteria in relation to CRC. Parabacteroides distasonis (Pa.d.) has been reported abundant in mice’s stools and inversely associated with intestinal tumor burden and IL-1β concentrations [26]. In their report, the Authors attributed to Pa.d. anti-inflammatory and anti-cancer properties that are likely mediated by the suppression of TLR4 and Akt signaling, as well as promotion of apoptosis [27].
CRC development has been associated with increased presence of Bacteroides fragilis (B.f.) in gut microbiota [24]. This Gram-negative, facultative anaerobe specie constitutes an appreciable proportion of the human gastrointestinal tract microbiota and, as typical of most gram-negative bacilli, it secretes an unusually complex mixture of neurotoxins, including the extremely pro-inflammatory lipopolysaccharide [11]. In this regard, it is important to underline the presence of the Gram+ Pediococcus pentasaceus (Pe.p.) in fecal samples of control subjects since this specie is able to eliminate the lipopolysaccharide (LPS) components, which can cause intestinal inflammation [28], that is a biological condition known to predispose to CRC development [29,30]. Furthermore, Pediococcus pentasaceus, found only in controls, showed antiproliferative activity on colon cancer cells directly adhering to those and triggering the bioproduction of short-chain fatty acids (SCFAs), mainly butyric and propionic acids, suggesting for the use of this bacteria specie as an alternative bio-prophylactic and biotherapeutic strategy for colon cancer [31]. Finally, we detected B. fragilis only in subjects with endoscopic pre-cancerous lesions (SCA/P).
What metabolic situation can derive from a shift of gut microbiota or, in general, by the induction of dysbiosis? From a biochemical point of view, dysbiosis has some common characteristics: the number of bacteria that produce SCFAs is reduced. This condition is negative because the SCFA strengthen the intestinal barrier and the immune system in the defense against pathogens. At the same time, levels of harmful microorganisms rise; these include bacteria, mostly Gram-negative bacteria, that lysing lipopolysaccharide (LPS) in the gut lumen facilitate its entry into bloodstream and may lead to a deleterious pro-inflammatory systemic immune response [32].
It is also known that dysbiosis leads to an increase of bacterial populations that stimulate tumorigenesis and contribute to epithelial carcinogenesis and tumor progression by altering metabolic properties (such as bile acid and butyric acid) and inflammatory process. In many human intestinal diseases, such as Crohn’s disease and ulcerative colitis, significant changes in gut microbiota have been reported [33,34,35]. In these pathological conditions, as well in CRC patients [4], again, one of the major biological effects observed is a reduction/substitution of butyrate-producing bacteria. Butyrate, one of the SCFAs, is a major metabolite in colonic lumen arising from bacterial fermentation of dietary fiber and has been shown to be a critical mediator of the colonic inflammatory response. Butyrate has been implicated in the protection against colitis and CRC by reducing oxidative damage to DNA, inducing apoptosis in DNA-damaged cells, inhibiting tumor cell growth, and decreasing the activity of co-carcinogenic enzymes [36,37]. Butyrate possesses both preventive and therapeutic potential to counteract inflammation-mediated ulcerative colitis (UC) and colorectal cancer. Emerging data suggest that certain groups of bacteria might promote whereas others might protect against colon cancer. Indeed, Fusobacterium nucleatum seems to play a central role in the tumor microenvironment as its abundance correlates with cancer progression as well as the dysbiotic tumor microbiota composition including Bacteroides spp, the enterotoxigenic Bacteroides fragilis in particular, and Prevotella species, may also promote colorectal cancer by stimulating exaggerated immune responses via Th17 cells [38]. Andersen and coworkers reported an increase of acetic and butyric acids in colon contents of pigs, when a commercially available probiotic strain Pediococcus pentosaceus was administrated [39], a Gram+ coccus again present only in fecal samples of control subjects.
Concerning the greater presence of Bacteroides vulgatus and Bacteroides uniformis in control stool samples compared to SCA/P subjects, it is worth mentioning the findings by Wang, which studied the discrepancies of gut microbiota between healthy subjects and CRC patients, reporting a structural segregation between the two populations [4]. In his report, Wang identified forty-eight operational taxonomic units (OTUs) by redundancy analysis as key variables significantly associated with the structural difference. One OTU closely related to Bacteroides fragilis (98.1% similarity) was enriched in the gut microbiota of CRC patients, whereas two OTUs related to Bacteroides vulgatus (100% and 98.7% similarity, respectively) and one to Bacteroides uniformis (97.5% similarity) was enriched in that of healthy volunteers. Revisiting our data, it is clear that Bacteroides fragilis is present only in the fecal samples of the SCA/P subjects, while Bacteroides vulgatus and Bacteroides uniformis are predominant in the feces of control subjects confirming, in part, the data coming from Wang’s report [4].
Several authors suggest that an altered composition in the gut microbiota, including the loss of microbiota diversity (LOMD), determines a predisposing microenvironment for dysbiosis. LOMD appears as a common feature of most dysbioses and it is a constant finding of intestinal dysbiosis [40]. In digestive diseases such as Crohn’s disease [41,42], irritable bowel syndrome, with [43] or without diarrhea [44] and colorectal cancer [45], LOMD is constantly observed. The duodenum-associated microbiota of celiac disease patients is also less diversified as previously reported also by our group [46,47]. According to this hypothesis, the LOMD may lead to an unstable microbial ecosystem favoring the emergence of dysbiotic microbiota and giving rise to immunological or metabolic diseases.

5. Conclusions

In conclusion, MALDI-TOF MS is a tool for rapid, accurate, and cost-effective identification of cultured bacteria and fungi in clinical microbiology. We agree with literature data suggesting that there is no single causative bacterial organism responsible in CRC. However, we believe that there is sufficient evidence suggesting that the reduction of protective bacteria or, vice versa, an increase of noxious strains, induced by diet in association with lifestyle and age, could influence the development of proliferative lesions. Apart from SCFAs produced by gut bacteria, the control of pathogenic species in the intestine could be determined by natural dietary compounds (i.e., polyphenols, essential oils, etc.) and a lot of immune regulators and mediators, partly tested in daily practice [48,49,50].
Future studies will enable to better understand both pro-carcinogenic and anti-carcinogenic mechanisms and will shed lights on the rational manipulation of the human microbiota and on the use of prebiotics, probiotics, or dietary modifications to improve health conditions.
The role of the diet in influencing the composition of the human microbiota is certainly not to be neglected. It is worldwide accepted that diet can influence the composition of the human microbiota, and that the supplementation with specific dietary resistant starch (RS) may lead to changes in fecal microbiota profiles associated with improved bowel health [51].
We know for sure that bacterial species might be greatly influenced by the presence of different nutrients, fibers etc. in diet and this aspect need to be resolved together with the concept of high variability and poor number of subjects included in the study.
Having this in mind, the manipulation of oncogenic, high risk gut microbiota may became an effective tool for cancer prevention, as well as treatment possibly based on fecal transplantation and lifestyle modifications.
However, this study was aimed to establish how different is the microbiota in subjects with or without intestinal polyps, regardless of dietary habits. So, for this study we just asked to the participants if they have had always the same dietary habits.
An evaluation of dietary habits would have made sense if we wanted to establish the type of foods that promote the development of certain bacterial species. But this aspect was beyond the scope of our study. In addition, although BMI was comparable between the two groups, no information is provided concerning the dietary habits in the weeks before assessment of intestinal microbiota.

Ethical Statement

The protocol study was approved by the Ethics Committee of the University of Bari, Protocol Record Policlinic Hospital, Bari, ClinicalTrials.gov Identifier: NCT03417258. Written informed consent was obtained from all study subjects.

Author Contributions

Conceptualization, L.P., M.B. and L.S.; Methodology, L.P., M.B. and L.S.; validation, L.P., M.B. and A.M.; investigation, L.P., M.B., A.M. and L.S.; resources, A.D.L.; data curation, M.B., M.T.V., and L.S.; writing—original draft preparation, L.P., M.B. and L.D.; writing—review and editing, L.P., M.B. and L.S.; visualization, M.B. and M.T.; supervision, A.D.L.; project administration, L.P., M.B. and L.S; funding acquisition, M.B. and A.D.

Funding

This paper has been partly funded by the Italian Ministry of University and Research—PON-BIOMIS 2018 (Project: Costituzione della Biobanca del Microbiota intestinale e salivare umano: dalla disbiosi alla simbiosi).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jemal, A.; Bray, F.; Center, M.M.; Ferlay, J.; Ward, E.; Forman, D. Global cancer statistics. CA Cancer J. Clin. 2011, 61, 69–90. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Santacroce, L.; Cagiano, R.; Del Prete, R.; Bottalico, L.; Sabatini, R.; Carlaio, R.G.; Prejbeanu, R.; Vermesan, H.; Dragulescu, S.I.; Vermesan, D.; et al. Helicobacter pylori infection and gastric MALTomas: An up-to-date and therapy highlight. Clin. Ter. 2008, 159, 457–462. [Google Scholar] [PubMed]
  3. Salyers, A.A. Bacteroides of the human lower intestinal tract. Annu. Rev. Microbiol. 1984, 38, 293–313. [Google Scholar] [CrossRef] [PubMed]
  4. Wang, T.; Cai, G.; Qiu, Y.; Fei, N.; Zhang, M.; Pang, X.; Jia, W.; Cai, S.; Zhao, L. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 2012, 6, 320–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. De Filippo, C.; Cavalieri, D.; Di Paola, M.; Ramazzotti, M.; Poullet, J.B.; Massart, S.; Collini, S.; Pieraccini, G.; Lionetti, P. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proc. Natl. Acad. Sci. USA 2010, 107, 14691–14696. [Google Scholar] [CrossRef] [Green Version]
  6. Walker, A.W.; Ince, J.; Duncan, S.H.; Webster, L.M.; Holtrop, G.; Ze, X.; Brown, D.; Stares, M.D.; Scott, P.; Bergerat, A.; et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J. 2010, 5, 220–230. [Google Scholar] [CrossRef]
  7. Zhang, C.H.; Zhang, M.H.; Wang, S.Y.; Han, R.J.; Cao, Y.F.; Hua, W.Y. Interactions between gut microbiota, host genetics and diet relevant to development of metabolic syndromes in mice. ISME J. 2010, 4, 232–241. [Google Scholar] [CrossRef] [Green Version]
  8. van Hylckama Vlieg, J.E.; Veiga, P.; Zhang, C.; Derrien, M.; Zhao, L. Impact of microbial transformation of food on health-from fermented foods to fermentation in the gastro-intestinal tract. Curr. Opin. Biotechnol. 2011, 22, 211–219. [Google Scholar] [CrossRef]
  9. Davis, C.D.; Milner, J.A. Gastrointestinal microflora, food components and colon cancer prevention. J. Nutr. Biochem. 2009, 20, 743–752. [Google Scholar] [CrossRef] [Green Version]
  10. Saleh, M.; Trinchieri, G. Innate immune mechanisms of colitis and colitis-associated colorectal cancer. Nat. Rev. Immunol. 2011, 11, 9–20. [Google Scholar] [CrossRef]
  11. Zhao, Y.; Lukiw, W.J. Bacteroidetes Neurotoxins and Inflammatory Neurodegeneration. Mol. Neurobiol. 2018, 55, 9100–9107. [Google Scholar] [CrossRef] [PubMed]
  12. Khosravi, A.; Mazmanian, S.K. Disruption of the gut microbiome as a risk factor for microbial infections. Curr. Opin. Microbiol. 2013, 16, 221–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Akin, H.; Tözün, N. Diet, microbiota, and colorectal cancer. J. Clin. Gastroenterol. 2014, 48 (Suppl. 1), S67–S69. [Google Scholar] [CrossRef] [PubMed]
  14. Shen, X.J.; Rawls, J.F.; Randall, T.; Burcal, L.; Mpande, C.N.; Jenkins, N.; Jovov, B.; Abdo, Z.; Sandler, R.S.; Keku, T.O. Molecular characterization of mucosal adherent bacteria and associations with colorectal adenomas. Gut Microbes 2010, 1, 138–147. [Google Scholar] [CrossRef] [Green Version]
  15. Barone, M.; Lofano, K.; De Tullio, N.; Licinio, R.; Albano, F.; Di Leo, A. Dietary, endocrine, and metabolic factors in the development of colorectal cancer. J. Gastrointest. Cancer 2012, 43, 13–19. [Google Scholar] [CrossRef]
  16. Schubert, S.; Kostrzewa, M. MALDI-TOF MS in the Microbiology Laboratory: Current Trends. Curr. Issues Mol. Biol. 2017, 23, 17–20. [Google Scholar] [CrossRef] [Green Version]
  17. Angeletti, S. Matrix assisted laser desorption time of flight mass spectrometry (MALDI-TOF MS) in clinical microbiology. J. Microbiol. Methods 2017, 138, 20–29. [Google Scholar] [CrossRef]
  18. Ballini, A.; Gnoni, A.; De Vito, D.; Dipalma, G.; Cantore, S.; Gargiulo Isacco, C.; Saini, R.; Santacroce, L.; Topi, S.; Scarano, A.; et al. Effect of probiotics on the occurrence of nutrition absorption capacities in healthy children: A randomized double-blinded placebo-controlled pilot study. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 8645–8657. [Google Scholar] [CrossRef]
  19. Campanella, V.; Syed, J.; Santacroce, L.; Saini, R.; Ballini, A.; Inchingolo, F. Oral probiotics influence oral and respiratory tract infections in pediatric population: A randomized double-blinded placebo-controlled pilot study. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 8034–8041. [Google Scholar]
  20. Ballini, A.; Santacroce, L.; Cantore, S.; Bottalico, L.; Dipalma, G.; Vito, D.; Saini, R.; Inchingolo, F. Probiotics Improve Urogenital Health in Women. Open Access Maced. J. Med. Sci. 2018, 6, 1845–1850. [Google Scholar] [CrossRef] [Green Version]
  21. Inchingolo, F.; Dipalma, G.; Cirulli, N.; Cantore, S.; Saini, R.S.; Altini, V.; Santacroce, L.; Ballini, A.; Saini, R. Microbiological results of improvement in periodontal condition by administration of oral probiotics. J. Biol. Regul. Homeost. Agents 2018, 32, 1323–1328. [Google Scholar] [PubMed]
  22. Ballini, A.; Santacroce, L.; Cantore, S.; Bottalico, L.; Dipalma, G.; Topi, S.; Saini, R.; De Vito, D.; Inchingolo, F. Probiotics Efficacy on Oxidative Stress Values in Inflammatory Bowel Disease: A Randomized Double-Blinded Placebo-Controlled Pilot Study. Endocr. Metab. Immune Disord. Drug Targets 2019, 19, 373–381. [Google Scholar] [CrossRef] [PubMed]
  23. Santacroce, L.; Charitos, I.A.; Bottalico, L. A successful history: Probiotics and their potential as antimicrobials. Expert Rev. Anti-Infect. Ther. 2019, 17, 635–645. [Google Scholar] [CrossRef] [PubMed]
  24. Zhu, Q.; Gao, R.; Wu, W.; Qin, H. The role of gut microbiota in the pathogenesis of colorectal cancer. Tumour Biol. 2013, 34, 1285–1300. [Google Scholar] [CrossRef] [PubMed]
  25. Kwong, T.N.Y.; Wang, X.; Nakatsu, G.; Chow, T.C.; Tipoe, T.; Dai, R.Z.W.; Tsoi, K.K.K.; Wong, M.C.S.; Tse, G.; Chan, M.T.V.; et al. Association between Bacteremia from Specific Microbes and Subsequent Diagnosis of Colorectal Cancer. Gastroenterology 2018, 155, 383–390. [Google Scholar] [CrossRef]
  26. Farhana, L.; Antaki, F.; Murshed, F.; Mahmud, H.; Judd, S.L.; Nangia-Makker, P.; Levi, E.; Yu, Y.; Majumdar, A.P. Gut microbiome profiling and colorectal cancer in African Americans and Caucasian Americans. World J. Gastrointest. Pathophysiol. 2018, 9, 47–58. [Google Scholar] [CrossRef]
  27. Koh, G.Y.; Kane, A.; Lee, K.; Xu, Q.; Wu, X.; Roper, J.; Mason, J.B.; Crott, J.W. Parabacteroides distasonis attenuates toll-like receptor 4 signaling and Akt activation and blocks colon tumor formation in high-fat diet-fed azoxymethane-treated mice. Int. J. Cancer 2018, 143, 1797–1805. [Google Scholar] [CrossRef] [Green Version]
  28. Kanauchi, M.; Kondo, A.; Asami, K. Eliminating Lipopolysaccharide (LPS) Using Lactic Acid Bacteria (LAB) and a Fraction of its LPS-Elimination Protein. Methods Mol. Biol. 2019, 1887, 167–174. [Google Scholar]
  29. Kashyap, D.; Garg, V.K.; Tuli, H.S.; Yerer, M.B.; Sak, K.; Sharma, A.K.; Kumar, M.; Aggarwal, V.; Sandhu, S.S. Fisetin and Quercetin: Promising Flavonoids with Chemopreventive Potential. Biomolecules 2019, 9, 174. [Google Scholar] [CrossRef] [Green Version]
  30. Strum, W.B.; Boland, C.R. Clinical and Genetic Characteristics of Colorectal Cancer in Persons under 50 Years of Age: A Review. Dig. Dis. Sci. 2019, 64, 3059–3065. [Google Scholar] [CrossRef]
  31. Thirabunyanon, M.; Hongwittayakorn, P. Potential probiotic lactic acid bacteria of human origin induce antiproliferation of colon cancer cells via synergic actions in adhesion to cancer cells and short-chain fatty acid bioproduction. Appl. Biochem. Biotechnol. 2013, 169, 511–525. [Google Scholar] [CrossRef] [PubMed]
  32. Gnauck, A.; Lentle, R.G.; Kruger, M.C. The Characteristics and Function of Bacterial Lipopolysaccharides and Their Endotoxic Potential in Humans. Int. Rev. Immunol. 2016, 35, 189–218. [Google Scholar] [CrossRef] [PubMed]
  33. Frank, D.N.; St Amand, A.L.; Feldman, R.A.; Boedeker, E.C.; Harpaz, N.; Pace, N.R. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl. Acad. Sci. USA 2007, 104, 13780–13785. [Google Scholar] [CrossRef] [Green Version]
  34. Willing, B.; Dicksved, J.; Halfvarson, J.; Andersson, A.; Lucio, M.; Zheng, Z.; Järnerot, G.; Tysk, C.; Jansson, J.K.; Engstrand, L. A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology 2010, 139, 1844–1854. [Google Scholar] [CrossRef] [PubMed]
  35. Willing, B.; Halfvarson, J.; Dicksved, J.; Rosenquist, M.; Jarnerot, G.; Engstrand, L.; Tysk, C.; Jansson, J.K. Twin studies reveal specific imbalances in the mucosa-associated microbiota of patients with ileal Crohn’s disease. Inflamm. Bowel Dis. 2009, 15, 653–660. [Google Scholar] [CrossRef] [PubMed]
  36. Pryde, S.E.; Duncan, S.H.; Hold, G.L.; Stewart, C.S.; Flint, H.J. The microbiology of butyrate formation in the human colon. FEMS Microbiol. Lett. 2002, 217, 133–139. [Google Scholar] [CrossRef] [PubMed]
  37. Rose, D.J.; DeMeo, M.T.; Keshavarzian, A.; Hamaker, B.R. Influence of dietary fiber on inflammatory bowel disease and colon cancer: Importance of fermentation pattern. Nutr. Rev. 2007, 65, 51–62. [Google Scholar] [CrossRef]
  38. Shiotani, A.; Fukushima, S.; Matsumoto, H. Carcinogenesis and Gut Microbiota. Gan Kagaku Ryoho 2019, 46, 199–204. [Google Scholar]
  39. Andersen, A.D.; Cilieborg, M.S.; Lauridsen, C.; Mørkbak, A.L.; Sangild, P.T. Supplementation with Lactobacillus paracasei or Pediococcus pentosaceus does not prevent diarrhoea in neonatal pigs infected with Escherichia coli F18. Br. J. Nutr. 2017, 118, 109–120. [Google Scholar] [CrossRef] [Green Version]
  40. Mosca, A.; Leclerc, M.; Hugot, J.P. Gut Microbiota Diversity and Human Diseases: Should We Reintroduce Key Predators in Our Ecosystem? Front. Microbiol. 2016, 7, 455. [Google Scholar] [CrossRef] [Green Version]
  41. Sha, S.; Xu, B.; Wang, X.; Zhang, Y.; Wang, H.; Kong, X.; Zhu, H.; Wu, K. The biodiversity and composition of the dominant fecal microbiota in patients with inflammatory bowel disease. Diagn. Microbiol. Infect. Dis. 2013, 75, 245–251. [Google Scholar] [CrossRef] [PubMed]
  42. Matsuoka, K.; Kanai, T. The gut microbiota and inflammatory bowel disease. Semin. Immunopathol. 2015, 37, 47–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Carroll, I.M.; Ringel-Kulka, T.; Siddle, J.P.; Ringel, Y. Alterations in composition and diversity of the intestinal microbiota in patients with diarrhea predominant irritable bowel syndrome. Neurogastroenterol. Motil. 2012, 24, 521–530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Durbán, A.; Abellán, J.J.; Jiménez-Hernández, N.; Salgado, P.; Ponce, M.; Ponce, J.; Garrigues, V.; Latorre, A.; Moya, A. Structural alterations of faecal and mucosa-associated bacterial communities in irritable bowel syndrome: Microbial diversity in irritable bowel syndrome. Environ. Microbiol. Rep. 2012, 4, 242–247. [Google Scholar]
  45. Ahn, J.; Sinha, R.; Pei, Z.; Dominianni, C.; Wu, J.; Shi, J.; Goedert, J.J.; Hayes, R.B.; Yang, L. Human gut microbiome and risk for colorectal cancer. J. Natl. Cancer Inst. 2013, 105, 1907–1911. [Google Scholar] [CrossRef] [Green Version]
  46. Schippa, S.; Iebba, V.; Barbato, M.; Di Nardo, G.; Totino, V.; Checchi, M.P.; Longhi, C.; Maiella, G.; Cucchiara, S.; Conte, M.P. A distinctive “microbial signature” in celiac pediatric patients. BMC Microbiol. 2010, 10, 175. [Google Scholar] [CrossRef]
  47. Francavilla, R.; Piccolo, M.; Francavilla, A.; Polimeno, L.; Semeraro, F.; Cristofori, F.; Castellaneta, S.; Barone, M.; Indrio, F.; Gobbetti, M.; et al. Clinical and Microbiological Effect of a Multispecies Probiotic Supplementation in Celiac Patients With Persistent IBS-type Symptoms: A Randomized, Double-Blind, Placebo-controlled, Multicenter Trial. J. Clin. Gastroenterol. 2019, 53, e117. [Google Scholar] [CrossRef] [Green Version]
  48. Man, A.; Santacroce, L.; Jacob, R.; Mare, A.; Man, L. Antimicrobial Activity of Six Essential Oils Against a Group of Human Pathogens: A Comparative Study. Pathogens 2019, 8, 15. [Google Scholar] [CrossRef] [Green Version]
  49. Ballini, A.; Cantore, S.; Saini, R.; Pettini, F.; Fotopoulou, E.A.; Saini, S.R.; Georgakopoulos, I.P.; Dipalma, G.; Gargiulo Isacco, C.; Inchingolo, F. Effect of activated charcoal probiotic toothpaste containing Lactobacillus paracasei and xylitol on dental caries: A randomized and controlled clinical trial. J. Biol. Regul. Homeost. Agents 2019, 33, 977–981. [Google Scholar]
  50. Di Serio, F.; Lovero, R.; D’Agostino, D.; Nisi, L.; Miragliotta, G.; Contino, R.; Man, A.; Ciccone, M.M.; Santacroce, L. Evaluation of procalcitonin, Vitamin D and C-reactive protein levels in septic patients with positive emocoltures. Our preliminary experience. Acta Med. Mediterr. 2016, 32, 1911–1914. [Google Scholar] [CrossRef]
  51. Maier, T.V.; Lucio, M.; Lee, L.H.; VerBerkmoes, N.C.; Brislawn, C.J.; Bernhardt, J.; Lamendella, R.; McDermott, J.E.; Bergeron, N.; Heinzmann, S.S.; et al. Impact of Dietary Resistant Starch on the Human Gut Microbiome, Metaproteome, and Metabolome. MBio 2017, 8, e01343-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Gut microbiota species recognized in the fecal samples of the control and sporadic colorectal adenomas/polyp (SCA/P) subjects: (a) Gram−bacteria; (b) Gram+ bacteria.
Figure 1. Gut microbiota species recognized in the fecal samples of the control and sporadic colorectal adenomas/polyp (SCA/P) subjects: (a) Gram−bacteria; (b) Gram+ bacteria.
Applsci 09 05491 g001
Table 1. Demographical and anthropometric parameters of the patients involved in the study.
Table 1. Demographical and anthropometric parameters of the patients involved in the study.
Pts. DataSCA/PControlp Value
Age63.4 ± 864.5 ± 40.56
SexM:14, F:6M:11, F:90.61
BMI29.8 ± 5.430.3 ± 7.20.76
Weight (Kgs)80.5 ± 12.377.3 ± 13.80.45
Table 2. Fecal anaerobic bacteria species isolated from the subjects involved in the study.
Table 2. Fecal anaerobic bacteria species isolated from the subjects involved in the study.
Gram+ BacteriaGram− Bacteria
Bifidobacterium sppBiff.Bacteroides fragilisB.f.
Clostridium clostridiformeC.c.Bacteroides ovatusB.o.
Clostridium innocuumC.i.Bacteroides stercorisB.s.
Clostridium ramosumC.r.Bacteroides thetaiotaomicronB.t.
Clostridium sardelliC.s.Bacteroides uniformisB.u.
Corynebacterium xerosusCor. x.Bacteroides vulgatusB.v.
Propionibacterium avidumPr.a.Prevotella melaninigenicaPre.m.
Eubacterium limosumEu.l.Parabacteroides distasonisPa.d.
Pediococcus pentosaceusPe.p.Parabacteroides merdaePa.m.

Share and Cite

MDPI and ACS Style

Polimeno, L.; Barone, M.; Mosca, A.; Viggiani, M.T.; Di Leo, A.; Debellis, L.; Troisi, M.; Daniele, A.; Santacroce, L. Gut Microbiota Imbalance is Related to Sporadic Colorectal Neoplasms. A Pilot Study. Appl. Sci. 2019, 9, 5491. https://doi.org/10.3390/app9245491

AMA Style

Polimeno L, Barone M, Mosca A, Viggiani MT, Di Leo A, Debellis L, Troisi M, Daniele A, Santacroce L. Gut Microbiota Imbalance is Related to Sporadic Colorectal Neoplasms. A Pilot Study. Applied Sciences. 2019; 9(24):5491. https://doi.org/10.3390/app9245491

Chicago/Turabian Style

Polimeno, Lorenzo, Michele Barone, Adriana Mosca, Maria Teresa Viggiani, Alfredo Di Leo, Lucantonio Debellis, Marco Troisi, Antonella Daniele, and Luigi Santacroce. 2019. "Gut Microbiota Imbalance is Related to Sporadic Colorectal Neoplasms. A Pilot Study" Applied Sciences 9, no. 24: 5491. https://doi.org/10.3390/app9245491

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop