The power of small changes: Comprehensive analyses of microbial dysbiosis in breast cancer

Sheetal Parida, Dipali Sharma, Sheetal Parida, Dipali Sharma

Abstract

Disparate occurrence of breast cancer remains an intriguing question since only a subset of women with known risk factors develop cancer. Recent studies suggest an active role of local and distant microbiota in breast cancer initiation, progression, and overall prognosis. A dysbiotic microbiota predisposes the body to develop cancer by inducing genetic instability, initiating DNA damage and proliferation of the damaged progeny, eliciting favorable immune response, metabolic dysregulation and altered response to therapy. In this review, we present our analyses of the existing datasets and discuss the local dysbiosis observed in breast cancer patients and different aspects of breast carcinogenesis that can be potentially influenced by local breast microbiota. Striking differences between microbial community compositions in breast of cancer patients compared to healthy individuals were noted. Differences in microbiome were also apparent between benign and malignant disease and between nipple aspirate fluid of healthy individuals and breast survivors. We also discuss the identification of distinct bacterial, fungal, viral as well as parasite signatures for breast cancer. These microbes are capable of producing numerous secondary metabolites that can act as signaling mediators effecting breast cancer progression. We review how microbes potentially alter response to therapy affecting drug metabolism, pharmacokinetics, anti-tumor effects and toxicity. In conclusion, breast harbors a community of microbes that can communicate with the host cells inducing downstream signaling pathways and modulating various aspects of breast cancer growth and metastatic progression and an improved understanding of microbial dysbiosis can potentially reduce breast cancer risk and improve outcomes of breast cancer patients. The human microbiome, now referred to as, the "forgotten organ" contains a metagenome that is 100-fold more diverse compared to the human genome, thereby, is critically associated with human health [1,2]. With the revelations of the human microbiome project and advent of deep sequencing techniques, a plethora of information has been acquired in recent years. Body sites like stomach, bladder and lungs, once thought to be sterile, are now known to harbor millions of indigenous microbial species. Approximately 80% of the healthy microbiome consists of Firmicutes and Bacteroidetes accompanied by Verrucomicrobia, Actinobacteria, Proteobacteria, Tenericutes and Cyanobacteria [2-7]. The role of microbiome in diabetes, obesity and even neurodegenerative diseases was greatly appreciated in the last decade [1,7-14] and now it has been established that microbiome significantly contributes to many organ specific cancers [1,15,16].

Conflict of interest statement

Disclosure statement

All authors declare that they have no conflict of interest.

Copyright © 2019 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Fig. 1.
Fig. 1.
A schematic representation of the planned analyses. Patient data from previous studies were analyzed to define common trends among their results as well as unique aspects using One Codex (a bioinformatics platform for microbial genomics).
Fig. 2.
Fig. 2.
(A) Relative abundance of major phyla. (B) Percentage of three major phyla, Firmicutes, Bacteroides and Proteobacteria. (C) Relative percentage of lactobacillus species in breast cancer compared to control. (D) Frequency of occurrence of abundant taxa in control and cancer tissue samples (SRP076038, Analysis: One Codex).
Fig. 3.
Fig. 3.
(A) Phylum distribution of local breast microbiota in cancer and nearby normal tissue samples. (B) Mean percentages of major phyla in cancer and nearby normal breast tissue samples. (C) Percentage of three major phyla, Firmicutes, Bacteroides and Proteobacteria. (D) Frequency of occurrence of abundant taxa in breast cancer and nearby normal tissue samples (PRJEB4755, Analysis: One Codex).
Fig. 4.
Fig. 4.
(A) Phylum distribution of breast ductal microbiota in Nipple aspirate fluid of breast cancer survivors and healthy volunteers. (B) Mean percentages of major phyla in nipple aspirate fluid of breast cancer survivors and healthy volunteers. (C) Percentage of three major phyla, Firmicutes, Bacteroides and Proteobacteria. (D) Frequency of occurrence of abundant taxa in NAF of breast cancer survivors and healthy volunteers (SRP071608, Analysis: One Codex).
Fig. 5.
Fig. 5.
(A) Phylum distribution in local breast microbiota of breast cancer patients having benign and malignant disease. (B) Mean percentages of major phyla in benign and malignant breast cancer tissue samples. (C) Percentage of three major phyla, Firmicutes, Bacteroides and Proteobacteria. (D) Frequency of occurrence of abundant taxa in benign and malignant breast tissue samples (PRJNA335375, EBI-ENA, Analysis: OneCodex).

References

    1. Schwabe RF, Jobin C, The microbiome and cancer, Nat. Rev. Cancer 13 (2013) 800.
    1. Plottel CS, Blaser MJ, Microbiome and malignancy, Cell Host Microbe 10 (2011) 324–335.
    1. Ohigashi S, Sudo K, Kobayashi D, et al., Changes of the intestinal microbiota, short chain fatty acids, and fecal pH in patients with colorectal cancer, Dig. Dis. Sci 58 (2013) 1717–1726.
    1. Ley RE, Obesity and the human microbiome, Curr. Opin. Gastroenterol 26 (2010) 5–11.
    1. Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI, Obesity alters gut microbial ecology, Proc. Natl. Acad. Sci. U. S. A 102 (2005) 11070–11075.
    1. Ley RE, Knight R, Gordon JI, The human microbiome: eliminating the biomedical/environmental dichotomy in microbial ecology, Environ. Microbiol 9 (2007) 3–4.
    1. Pevsner-Fischer M, Tuganbaev T, Meijer M, Zhang SH, Zeng ZR, Chen MH, Elinav E, Role of the microbiome in non-gastrointestinal cancers, World J. Clin. Oncol 7 (2016) 200–213.
    1. Abu-Shanab A, Quigley EM, The role of the gut microbiota in nonalcoholic fatty liver disease, Nat. Rev. Gastroenterol. Hepatol 7 (2010) 691.
    1. Contreras AV, Cocom-Chan B, Hernandez-Montes G, Portillo-Bobadilla T, Resendis-Antonio O, Host-microbiome interaction and cancer: potential application in precision medicine, Front. Physiol 7 (2016).
    1. Samuel BS, Shaito A, Motoike T, et al., Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41, Proc. Natl. Acad. Sci 105 (2008) 16767–16772.
    1. Dapito DH, Mencin A, Gwak GY, Pradere JP, Jang MK, Mederacke I, Caviglia JM, Khiabanian H, Adeyemi A, Bataller R, Lefkowitch JH, Bower M, Friedman R, Sartor RB, Rabadan R, Schwabe RF, Promotion of hepatocellular carcinoma by the intestinal microbiota and TLR4, Cancer Cell 21 (2012) 504–516.
    1. Phelan JP, Reen FJ, Caparros-Martin JA, O’Connor R, O’Gara F, Rethinking the bile acid/gut microbiome axis in cancer, Oncotarget. 8 (2017) 115,736–115,747.
    1. Vieira SM, Pagovich OE, Kriegel MA, Diet, microbiota and autoimmune diseases, Lupus. 23 (2014) 518–526.
    1. Parracho HM, Bingham MO, Gibson GR, McCartney AL, Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children, J. Med. Microbiol 54 (2005) 987–991.
    1. Vogtmann E, Goedert JJ, Epidemiologic studies of the human microbiome and cancer, Br. J. Cancer 114 (2016) 237.
    1. Yang J, Tan Q, Fu Q, et al., Gastrointestinal microbiome and breast cancer: correlations, mechanisms and potential clinical implications, Breast Cancer. 24 (2017) 220–228.
    1. Ma C, Han M, Heinrich B, et al., Gut microbiome–mediated bile acid metabolism regulates liver cancer via NKT cells, Science (2018) 360 eaan5931.
    1. Freudenheim JL, Genco RJ, LaMonte MJ, et al., Periodontal disease and breast cancer: prospective cohort study of postmenopausal women, Cancer Epidemiol. Biomark. Prev 25 (2016) 43–50.
    1. Sfreddo CS, Maier J, De David SC, Susin C, Moreira CHC, Periodontitis and breast cancer: a case-control study, Community Dent. Oral Epidemiol 45 (2017) 545–551.
    1. Nwizu NN, Marshall JR, Moysich K, et al., Periodontal disease and incident cancer risk among postmenopausal women: results from the Women’s Health Initiative Observational Cohort, Cancer Epidemiol. Biomark. Prev 26 (2017) 1255–1265.
    1. Mai X, LaMonte MJ, Hovey KM, et al., Periodontal disease severity and cancer risk in postmenopausal women: the Buffalo OsteoPerio study, Cancer Causes Control 27 (2016) 217–228.
    1. Wang H, Altemus J, Niazi F, Green H, Calhoun BC, Sturgis C, Grobmyer SR, Eng C, Breast tissue, oral and urinary microbiomes in breast cancer, Oncotarget. 8 (2017) 88,122–88,138.
    1. DeStefano Shields CE, Van Meerbeke SW, Housseau F, et al., Reduction of murine colon tumorigenesis driven by enterotoxigenic bacteroides fragilis using cefoxitin treatment, J. Infect. Dis 214 (2016) 122–129.
    1. Fernández L, Cárdenas N, Arroyo R, et al., Prevention of infectious mastitis by oral administration of lactobacillus salivarius PS2 during late pregnancy, Clin. Infect. Dis 62 (2016) 568–573.
    1. Pompei A, Cordisco L, Amaretti A, Zanoni S, Matteuzzi D, Rossi M, Folate production by bifidobacteria as a potential probiotic property, Appl. Environ. Microbiol 73 (2006) 179–185.
    1. Wollowski I, Rechkemmer G, Pool-Zobel BL, Protective role of probiotics and prebiotics in colon cancer, Am. J. Clin. Nutr 73 (2001) 451s–455s.
    1. Lakritz JR, Poutahidis T, Levkovich T, Varian BJ, Ibrahim YM, Chatzigiagkos A, Mirabal S, Alm EJ, Erdman SE, Beneficial bacteria stimulate host immune cells to counteract dietary and genetic predisposition to mammary cancer in mice, Int. J. Cancer 135 (2013) 529–540.
    1. Zhang W, Lu H, Graham DY, An update on helicobacter pylori as the cause of gastric cancer, Gastrointest. Tumors 1 (2014) 155–165.
    1. Ding SZ, Goldberg JB, Hatakeyama M, Helicobacter pylori infection, oncogenic pathways and epigenetic mechanisms in gastric carcinogenesis, Future Oncol. 6 (2010) 851–862.
    1. Backert S, Selbach M, Role of type IV secretion in Helicobacter pylori pathogenesis, Cell. Microbiol 10 (2008) 1573–1581.
    1. Lee IO, Kim JH, Choi YJ, Pillinger MH, Kim SY, Blaser MJ, Lee YC, Helicobacter pylori CagA phosphorylation status determines the gp130-activated SHP2/ERK and JAK/STAT signal transduction pathways in gastric epithelial cells, J. Biol. Chem 285 (2010) 16,042–16,050.
    1. Urbaniak C, Cummins J, Brackstone M, Macklaim JM, Gloor GB, Baban CK, Scott L, O’Hanlon DM, Burton JP, Francis KP, Tangney M, Reid G, Microbiota of human breast tissue, Appl. Environ. Microbiol 80 (2014) 3007–3014.
    1. Xuan C, Shamonki JM, Chung A, Dinome ML, Chung M, Sieling PA, Lee DJ, Microbial dysbiosis is associated with human breast cancer, PLoS ONE 9 (2014) e83744.
    1. Murphy EA, Velazquez KT, Herbert KM, Influence of high-fat diet on gut microbiota: a driving force for chronic disease risk, Curr. Opin. Clin. Nutr. Metab. Care 18 (2015) 515–520.
    1. Urbaniak C, Gloor GB, Brackstone M, Scott L, Tangney M, Reid G, The microbiota of breast tissue and its association with breast cancer, Appl. Environ. Microbiol 82 (2016) 5039–5048.
    1. Aragón F, Carino S, Perdigón G, de A Moreno de LeBlanc, Inhibition of growth and metastasis of breast cancer in mice by milk fermented with lactobacillus casei CRL 431, J. Immunother 38 (2015) 185–196.
    1. Marschalek J, Farr A, Marschalek ML, Domig KJ, Kneifel W, Singer CF, Kiss H, Petricevic L, Influence of orally administered probiotic lactobacillus strains on vaginal microbiota in women with breast cancer during chemotherapy: a randomized placebo-controlled double-blinded pilot study, Breast Care (Basel) 12 (2017) 335–339.
    1. Ojanotko-Harri A, Nikkari T, Harrl M-P, Paunio K, Metabolism of progesterone and testosterone by Bacillus cereus strain Socransky 67 and Streptococcus mutans strain Ingbritt, Oral Microbiol. Immunol 5 (1990) 237–239.
    1. Wiebe JP, Muzia D, Hu J, Szwajcer D, Hill SA, Seachrist JL, The 4-pregnene and 5alpha-pregnane progesterone metabolites formed in nontumorous and tumorous breast tissue have opposite effects on breast cell proliferation and adhesion, Cancer Res. 60 (2000) 936–943.
    1. Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C, Bielski C, Rostron T, Cerundolo V, Pamer EG, Abramson SB, Huttenhower C, Littman DR, Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis, Elife. 2 (2013) e01202.
    1. Chan AA, Bashir M, Rivas MN, Duvall K, Sieling PA, Pieber TR, Vaishampayan PA, Love SM, Lee DJ, Characterization of the microbiome of nipple aspirate fluid of breast cancer survivors, Sci. Rep 6 (2016) 28,061.
    1. Saulnier DM, Kevin R, Mistretta TA, Diaz MA, Mandal D, Raza S, Weidler EM, Qin X, Coarfa C, Milosavljevic A, Petrosino JF, Highlander S, Gibbs R, Lynch SV, Shulman RJ, Versalovic J, Gastrointestinal microbiome signatures of pediatric patients with irritable bowel syndrome, Gastroenterology. 141 (2011) 1782–1791.
    1. Martínez I, Muller CE, Walter J, Long-term temporal analysis of the human fecal microbiota revealed a stable core of dominant bacterial species, PLoS ONE 8 (2013) e69621.
    1. Clarke SF, Murphy EF, O’Sullivan O, Ross RP, O’Toole PW, Shanahan F, Cotter PD, Targeting the microbiota to address diet-induced obesity: a time dependent challenge, PLoS ONE 8 (2013) e65790.
    1. Hieken TJ, Chen J, Hoskin TL, et al., The microbiome of aseptically collected human breast tissue in benign and malignant disease, Sci. Rep 6 (2016) 30751.
    1. Hoffman RM, Development of recombinant methioninase to target the general cancer-specific metabolic defect of methionine dependence: a 40-year odyssey, Expert. Opin. Biol. Ther 15 (2015) 21–31.
    1. Cavuoto P, Fenech MF, A review of methionine dependency and the role of methionine restriction in cancer growth control and life-span extension, Cancer Treat. Rev 38 (2012) 726–736.
    1. Lakritz JR, Poutahidis T, Mirabal S, Varian BJ, Levkovich T, Ibrahim YM, Ward JM, Teng EC, Fisher B, Parry N, Lesage S, Alberg N, Gourishetti S, Fox JG, Ge Z, Erdman SE, Gut bacteria require neutrophils to promote mammary tumorigenesis, Oncotarget. 6 (2015) 9387–9396.
    1. Banerjee S, Wei Z, Tan F, et al., Distinct microbiological signatures associated with triple negative breast cancer, Sci. Rep 5 (2015) 15162.
    1. Banerjee S, Tian T, Wei Z, et al., Distinct Microbial Signatures Associated With Different Breast Cancer Types, (2018), p. 9.
    1. Baldwin DA, Feldman M, Alwine JC, Robertson ES, Metagenomic Assay for Identification of Microbial Pathogens in Tumor Tissues, 5 (2014), p. e01714.
    1. Ueda N, Wise LM, Stacker SA, Fleming SB, Mercer AA, Pseudocowpox virus encodes a homolog of vascular endothelial growth factor, Virology. 305 (2003) 298–309.
    1. Kristek J, Marjanović K, Dmitrović B, Krajinović Z, Sakić K, Trichinella spiralis and breast carcinoma–a case report, Coll Antropol. 29 (2005) 775–777.
    1. Benamrouz S, Conseil V, Creusy C, Calderon E, Dei-Cas E, Certad G, Parasites and malignancies, a review, with emphasis on digestive cancer induced by Cryptosporidium parvum (Alveolata: Apicomplexa: ), Parasite. 19 (2012) 101–115.
    1. Menon BS, Abdullah MS, Mahamud F, Singh B, Brief report. Intestinal parasites in Malaysian children with cancer, J. Trop. Pediatr 45 (1999) 241–242.
    1. Guarner J, Matilde-Nava T, Villaseñor-Flores R, Sanchez-Mejorada G, Frequency of intestinal parasites in adult cancer patients in Mexico, Arch. Med. Res 28 (1997) 219–222.
    1. Heussler VT, Küenzi P, Rottenberg S, Inhibition of apoptosis by intracellular protozoan parasites, Int. J. Parasitol 31 (2001) 1166–1176.
    1. Lowe SW, Lin AW, Apoptosis in cancer, Carcinogenesis. 21 (2000) 485–495.
    1. Roy S, Trinchieri G, Microbiota: a key orchestrator of cancer therapy, Nat. Rev. Cancer 17 (2017) 271.
    1. Touchefeu Y, Montassier E, Nieman K, et al., Systematic Review: The Role of the Gut Microbiota in Chemotherapy- or Radiation-Induced Gastrointestinal Mucositis – Current Evidence and Potential Clinical Applications, 40 (2014), pp. 409–421.
    1. Wang A, Ling Z, Yang Z, Kiela PR, Wang T, Wang C, Cao L, Geng F, Shen M, Ran X, Su Y, Cheng T, Wang J, Gut microbial dysbiosis may predict diarrhea and fatigue in patients undergoing pelvic cancer radiotherapy: a pilot study, PLoS ONE 10 (2015) e0126312.
    1. Haiser HJ, Turnbaugh PJ, Developing a metagenomic view of xenobiotic metabolism, Pharmacol. Res 69 (2012) 21–31.
    1. Selwyn FP, Cheng SL, Klaassen CD, Cui JY, Regulation of Hepatic Drug-Metabolizing Enzymes in Germ-Free Mice by Conventionalization and Probiotics, 44 (2016), pp. 262–274.
    1. Selwyn FP, Cui JY, Klaassen CD, RNA-Seq Quantification of Hepatic Drug Processing Genes in Germ-Free Mice, 43 (2015), pp. 1572–1580.
    1. Fujita K, Sparreboom A, Pharmacogenetics of irinotecan disposition and toxicity: a review, Curr. Clin. Pharmacol 5 (2010) 209–217.
    1. Lehouritis P, Cummins J, Stanton M, et al., Local bacteria affect the efficacy of chemotherapeutic drugs, Sci. Rep 5 (2015) 14554.
    1. Iida N, Dzutsev A, Stewart CA, et al., Commensal Bacteria Control Cancer Response to Therapy by Modulating the Tumor Microenvironment, 342 (2013), pp. 967–970.
    1. Hooper LV, Macpherson AJ, Immune adaptations that maintain homeostasis with the intestinal microbiota, Nat. Rev. Immunol 10 (2010) 159.
    1. Sonis ST, The pathobiology of mucositis, Nat. Rev. Cancer 4 (2004) 277.
    1. Viaud S, Saccheri F, Mignot G, et al., The Intestinal Microbiota Modulates the Anticancer Immune Effects of Cyclophosphamide, 342 (2013), pp. 971–976.
    1. Galluzzi L, Vitale I, Michels J, et al., Systems biology of cisplatin resistance: past, present and future, Cell Death Dis. 5 (2014) e1257.
    1. Kim S, Lee T-J, Park J-W, Kwon TK, Overexpression of cFLIPs Inhibits Oxaliplatin-Mediated Apoptosis Through Enhanced XIAP Stability and Akt Activation in Human Renal Cancer Cells, 105 (2008), pp. 971–979.
    1. Laurent A, Nicco C, Chéreau C, et al., Controlling Tumor Growth by Modulating Endogenous Production of Reactive Oxygen Species, 65 (2005), pp. 948–956.
    1. Gui QF, Lu HF, Zhang CX, Xu ZR, Yang YH, Well-balanced commensal microbiota contributes to anti-cancer response in a lung cancer mouse model, Genet. Mol. Res 14 (2015) 5642–5651.
    1. Tesniere A, Schlemmer F, Boige V, et al., Immunogenic death of colon cancer cells treated with oxaliplatin, Oncogene. 29 (2009) 482.
    1. Michaud M, Sukkurwala AQ, Martins I, Shen S, Zitvogel L, Kroemer G, Subversion of the chemotherapy-induced anticancer immune response by the ecto-ATPase CD39, OncoImmunology. 1 (2012) 393–395.
    1. Ghiringhelli F, Apetoh L, Tesniere A, et al., Activation of the NLRP3 inflammasome in dendritic cells induces IL-1β–dependent adaptive immunity against tumors, Nat. Med 15 (2009) 1170.
    1. Pateras IS, Havaki S, Nikitopoulou X, Vougas K, Townsend PA, Panayiotidis MI, Georgakilas AG, Gorgoulis VG, The DNA damage response and immune signaling alliance: is it good or bad? Nature decides when and where, Pharmacol. Ther 154 (2015) 36–56.
    1. Vacchelli E, Ma Y, Baracco EE, et al., Chemotherapy-Induced Antitumor Immunity Requires Formyl Peptide Receptor, 350 (2015), pp. 972–978.
    1. Daillère R, Vétizou M, Waldschmitt N, Yamazaki T, Isnard C, Poirier-Colame V, Duong CPM, Flament C, Lepage P, Roberti MP, Routy B, Jacquelot N, Apetoh L, Becharef S, Rusakiewicz S, Langella P, Sokol H, Kroemer G, Enot D, Roux A, Eggermont A, Tartour E, Johannes L, Woerther PL, Chachaty E, Soria JC, Golden E, Formenti S, Plebanski M, Madondo M, Rosenstiel P, Raoult D, Cattoir V, Boneca IG, Chamaillard M, Zitvogel L, Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects, Immunity 45 (2016) 931–943.
    1. Chitapanarux I, Chitapanarux T, Traisathit P, Kudumpee S, Tharavichitkul E, Lorvidhaya V, Randomized controlled trial of live Lactobacillus Acidophilus plus Bifidobacterium Bifidum in prophylaxis of diarrhea during radiotherapy in cervical cancer patients, 5 (2010), p. 31.
    1. Wang Y, Luo X, Pan H, Huang W, Wang X, Wen H, Shen K, Jin B, Pharmacological inhibition of NADPH oxidase protects against cisplatin induced nephrotoxicity in mice by two step mechanism, Food Chem. Toxicol 83 (2015) 251–260.
    1. Mercado-Lubo R, The interaction of gut microbes with host ABC transporters, Gut Microbes 1 (2010) 301–306.
    1. Cario E, Toll-like receptors in the pathogenesis of chemotherapy-induced gastrointestinal toxicity, Curr. Opin. Support. Palliat. Care 10 (2016) 157–164.
    1. Frank M, Hennenberg EM, Eyking A, et al., TLR Signaling Modulates Side Effects of Anticancer Therapy in the Small Intestine, 194 (2015), pp. 1983–1995.
    1. Napeñas JJ, Brennan MT, Coleman S, et al., Molecular methodology to assess the impact of cancer chemotherapy on the oral bacterial flora: a pilot study, Oral Surg. Oral Med. Oral Pathol. Oral Radiol. Endod 109 (2010) 554–560.
    1. Niu QY, Li ZY, Du GH, Qin XM, (1)H NMR based metabolomic profiling revealed doxorubicin-induced systematic alterations in a rat model, J. Pharm. Biomed. Anal 118 (2016) 338–348.
    1. Rigby RJ, Carr J, Orgel K, King SL, Lund PK, Dekaney CM, Intestinal bacteria are necessary for doxorubicin-induced intestinal damage but not for doxorubicin-induced apoptosis, Gut Microbes 7 (2016) 414–423.
    1. Nigro G, Rossi R, Commere PH, Jay P, Sansonetti PJ, The cytosolic bacterial peptidoglycan sensor Nod2 affords stem cell protection and links microbes to gut epithelial regeneration, Cell Host Microbe 15 (2014) 792–798.
    1. Jiang C, Xie C, Lv Y, et al., Intestine-selective farnesoid X receptor inhibition improves obesity-related metabolic dysfunction, Nat. Commun 6 (2015) 10166.
    1. Parséus A, Sommer N, Sommer F, et al., Microbiota-Induced Obesity Requires Farnesoid X Receptor, 66 (2017), pp. 429–437.
    1. Das SK, Eder S, Schauer S, et al., Adipose Triglyceride Lipase Contributes to Cancer-Associated Cachexia, 333 (2011), pp. 233–238.
    1. Ruud J, Wilhelms DB, Nilsson A, et al., Inflammation- and tumor-induced anorexia and weight loss require MyD88 in hematopoietic/myeloid cells but not in brain endothelial or neural cells, FASEB J. 27 (2013) 1973–1980.
    1. Suárez-Zamorano N, Fabbiano S, Chevalier C, et al., Microbiota depletion promotes browning of white adipose tissue and reduces obesity, Nat. Med 21 (2015) 1497.
    1. de Matos-Neto EM, Lima JD, de Pereira WO, et al., Systemic Inflammation in Cachexia – Is Tumor Cytokine Expression Profile the Culprit? Front. Immunol 6 (2015) 629.
    1. Bruggeman AR, Kamal AH, LeBlanc TW, Ma JD, Baracos VE, Roeland EJ, Cancer Cachexia: Beyond Weight Loss, 12 (2016), pp. 1163–1171.
    1. Cvan Trobec K, Kerec Kos M, Trontelj J, et al., Influence of Cancer Cachexia on Drug Liver Metabolism and Renal Elimination in Rats, 6 (2015), pp. 45–52.
    1. Bindels LB, Delzenne NM, Muscle wasting: the gut microbiota as a new therapeutic target? Int. J. Biochem. Cell Biol 45 (2013) 2186–2190.
    1. Klein GL, Petschow BW, Shaw AL, Weaver E, Gut barrier dysfunction and microbial translocation in cancer cachexia: a new therapeutic target, Curr. Opin. Support. Palliat. Care 7 (2013) 361–367.
    1. Schieber AM, Lee YM, Chang MW, Leblanc M, Collins B, Downes M, Evans RM, Ayres JS, Disease tolerance mediated by microbiome < em > E. coli < /em > involves inflammasome and IGF-1 signaling, 350 (2015), pp. 558–563.
    1. Apetoh L, Ghiringhelli F, Tesniere A, et al., Toll-like receptor 4–dependent contribution of the immune system to anticancer chemotherapy and radiotherapy, Nat. Med 13 (2007) 1050–1059.
    1. Ermolaeva MA, Segref A, Dakhovnik A, et al., DNA damage in germ cells induces an innate immune response that triggers systemic stress resistance, Nature. 501 (2013) 416.
    1. Al-Mayah A, Bright S, Chapman K, Irons S, Luo P, Carter D, Goodwin E, Kadhim M, The non-targeted effects of radiation are perpetuated by exosomes, Mutat. Res 772 (2015) 38–45.
    1. Demaria S, Ng B, Devitt ML, et al., Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated, Int. J. Radiat. Oncol. Biol. Phys 58 (2004) 862–870.
    1. Deng L, Liang H, Burnette B, Beckett M, Darga T, Weichselbaum RR, Fu YX, Irradiation and anti-PD-L1 treatment synergistically promote antitumor immunity in mice, J. Clin. Invest 124 (2014) 687–695.
    1. Baird JR, Friedman D, Cottam B, Dubensky TW, Kanne DB, Bambina S, Bahjat K, Crittenden MR, Gough MJ, Radiotherapy combined with novel STING-targeting oligonucleotides results in regression of established Tumors, Cancer Res. 76 (2015) 50–61.
    1. Barker HE, Paget JT, Khan AA, Harrington KJ, The tumor microenvironment after radiotherapy: mechanisms of resistance and recurrence, Nat. Rev. Cancer 15 (2015) 409–425.
    1. Vacchelli E, Eggermont A, Sautès-Fridman C, et al., Trial watch: Toll-like receptor agonists for cancer therapy, OncoImmunology. 2 (2013) e25238.
    1. Jones RM, Luo L, Ardita CS, et al., Symbiotic Lactobacilli Stimulate Gut Epithelial Proliferation < em > via < /em > Nox-Mediated Generation Of Reactive Oxygen Species, 32 (2013), pp. 3017–3028.
    1. Jones RM, Desai C, Darby TM, Luo L, Wolfarth AA, Scharer CD, Ardita CS, Reedy AR, Keebaugh ES, Neish AS, Lactobacilli modulate epithelial Cytoprotection through the Nrf2 pathway, Cell Rep. 12 (2015) 1217–1225.
    1. Blanarova C, Galovicova A, Petrasova D, Use of probiotics for prevention of radiation-induced diarrhea, Bratisl. Lek. Listy 110 (2009) 98–104.
    1. Sharma A, Rath GK, Chaudhary SP, Thakar A, Mohanti BK, Bahadur S, < em > Lactobacillus brevis < /em > CD2 lozenges reduce radiation- and chemotherapy-induced mucositis in patients with head and neck cancer: A randomized double-blind placebo-controlled study, Eur. J. Cancer 48 (2012) 875–881.
    1. Duncan AM, Ronen A, Blakey DH, Diurnal variation in the response of gamma-ray-induced apoptosis in the mouse intestinal epithelium, Cancer Lett. 21 (1983) 163–166.
    1. Ishihara H, Tanaka I, Yakumaru H, et al., Circadian transitions in radiation dose-dependent augmentation of mRNA levels for DNA damage-induced genes elicited by accurate real-time RT-PCR quantification, J. Radiat. Res 51 (2010) 265–275.
    1. Ruifrok AC, Weil MM, Thames HD, Mason KA, Diurnal variations in the expression of radiation-induced apoptosis, Radiat. Res 149 (1998) 360–365.
    1. Leone V, Gibbons SM, Martinez K, et al., Effects of diurnal variation of gut microbes and high-fat feeding on host circadian clock function and metabolism, Cell Host Microbe 17 (2015) 681–689.
    1. Liang X, Bushman FD, FitzGerald GA, Rhythmicity of the intestinal microbiota is regulated by gender and the host circadian clock, Proc. Natl. Acad. Sci. U. S. A 112 (2015) 10479–10484.
    1. Mukherji A, Kobiita A, Ye T, Chambon P, Homeostasis in intestinal epithelium is orchestrated by the circadian clock and microbiota cues transduced by TLRs, Cell. 153 (2013) 812–827.
    1. Nguyen KD, Fentress SJ, Qiu Y, Yun K, Cox JS, Chawla A, Circadian Gene Bmal1 Regulates Diurnal Oscillations of Ly6Chi Inflammatory Monocytes, 341 (2013), pp. 1483–1488.
    1. Thaiss CA, Zeevi D, Levy M, Zilberman-Schapira G, Suez J, Tengeler AC, Abramson L, Katz MN, Korem T, Zmora N, Kuperman Y, Biton I, Gilad S, Harmelin A, Shapiro H, Halpern Z, Segal E, Elinav E, Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis, Cell. 159 (2014) 514–529.
    1. Maier I, Berry DM, Schiestl RH, Intestinal microbiota reduces genotoxic endpoints induced by high-energy protons, Radiat. Res 181 (2014) 45–53.

Source: PubMed

3
購読する