Cargando…

The Oral Mouse Microbiome Promotes Tumorigenesis in Oral Squamous Cell Carcinoma

Oral squamous cell carcinoma (OSCC) is the most common malignancy of the head and neck worldwide. Dysbiosis of the microbiome has increasingly been linked to the development of different kinds of cancer. Applying 16S rRNA gene sequence analysis and metatranscriptomic analyses, we characterized the l...

Descripción completa

Detalles Bibliográficos
Autores principales: Stashenko, Philip, Yost, Susan, Choi, Yoonhee, Danciu, Theodora, Chen, Tsute, Yoganathan, Subbiah, Kressirer, Christine, Ruiz-Tourrella, Montserrat, Das, Bikul, Kokaras, Alexis, Frias-Lopez, Jorge
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society for Microbiology 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6687944/
https://www.ncbi.nlm.nih.gov/pubmed/31387932
http://dx.doi.org/10.1128/mSystems.00323-19
_version_ 1783442809977569280
author Stashenko, Philip
Yost, Susan
Choi, Yoonhee
Danciu, Theodora
Chen, Tsute
Yoganathan, Subbiah
Kressirer, Christine
Ruiz-Tourrella, Montserrat
Das, Bikul
Kokaras, Alexis
Frias-Lopez, Jorge
author_facet Stashenko, Philip
Yost, Susan
Choi, Yoonhee
Danciu, Theodora
Chen, Tsute
Yoganathan, Subbiah
Kressirer, Christine
Ruiz-Tourrella, Montserrat
Das, Bikul
Kokaras, Alexis
Frias-Lopez, Jorge
author_sort Stashenko, Philip
collection PubMed
description Oral squamous cell carcinoma (OSCC) is the most common malignancy of the head and neck worldwide. Dysbiosis of the microbiome has increasingly been linked to the development of different kinds of cancer. Applying 16S rRNA gene sequence analysis and metatranscriptomic analyses, we characterized the longitudinal changes in the profiles and the function of the oral microbiome in a 4-nitroquinoline-1-oxide (4-NQO)-induced model of OSCC in gnotobiotic mice. We characterized the dynamics of the oral microbiome in this model using two different microbiome inocula: one from healthy mice and the other from mice bearing a 4-NQO-induced tumor. Mice colonized with different oral microbiomes and exposed to 4-NQO had increased tumor numbers and sizes compared to controls exposed to 4-NQO but lacking a microbiome. We observed an overall increase in diversity in the tumorigenic samples compared to that in the nontumor group not exposed to 4-NQO. Despite the variability in community dynamics, specific patterns emerged during the progression of the disease. In the two groups that were inoculated with the OSCC-associated microbiome, we observed opposite profiles of abundance in Parabacteroides and Corynebacterium. While the percentage of Parabacteroides bacteria decreased in the control group, it increased in the OSCC group, and the opposite was observed for Corynebacterium. The metatranscriptomic analysis revealed overexpression of the same metabolic signatures associated with OSCC regardless of the community profile. These included nitrogen transport, response to stress, interspecies interactions, Wnt pathway modulation, and amino acid and lipid biosynthesis. Thus, these results seem to suggest that certain collective physiological activities are critical for microbiome-mediated OSCC progression. IMPORTANCE There is growing evidence that changes in the microbiome are associated with carcinogenesis. To date, no consistent oral microbiome composition associated with OSCC has been identified. Longitudinal and functional studies like the study presented here should yield a better understanding of the role that the oral microbiome plays in OSCC. Our findings, obtained using a germ-free mouse model, indicate that the presence of different oral microbiomes enhances tumorigenesis and increases the final number of tumors in mice. By studying community-wide expression profiles, we found that regardless of the phylogenetic composition of the microbiome, the same metabolic activities were consistently associated with OSCC. Therefore, due to the functional redundancy of the microbiome, the critical element in explaining the contribution of the microbiota in OSCC is the collective physiological activity of the community, thus accounting for the previous inability to identify a consensus community profile or etiologic agents for OSCC.
format Online
Article
Text
id pubmed-6687944
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher American Society for Microbiology
record_format MEDLINE/PubMed
spelling pubmed-66879442019-08-13 The Oral Mouse Microbiome Promotes Tumorigenesis in Oral Squamous Cell Carcinoma Stashenko, Philip Yost, Susan Choi, Yoonhee Danciu, Theodora Chen, Tsute Yoganathan, Subbiah Kressirer, Christine Ruiz-Tourrella, Montserrat Das, Bikul Kokaras, Alexis Frias-Lopez, Jorge mSystems Research Article Oral squamous cell carcinoma (OSCC) is the most common malignancy of the head and neck worldwide. Dysbiosis of the microbiome has increasingly been linked to the development of different kinds of cancer. Applying 16S rRNA gene sequence analysis and metatranscriptomic analyses, we characterized the longitudinal changes in the profiles and the function of the oral microbiome in a 4-nitroquinoline-1-oxide (4-NQO)-induced model of OSCC in gnotobiotic mice. We characterized the dynamics of the oral microbiome in this model using two different microbiome inocula: one from healthy mice and the other from mice bearing a 4-NQO-induced tumor. Mice colonized with different oral microbiomes and exposed to 4-NQO had increased tumor numbers and sizes compared to controls exposed to 4-NQO but lacking a microbiome. We observed an overall increase in diversity in the tumorigenic samples compared to that in the nontumor group not exposed to 4-NQO. Despite the variability in community dynamics, specific patterns emerged during the progression of the disease. In the two groups that were inoculated with the OSCC-associated microbiome, we observed opposite profiles of abundance in Parabacteroides and Corynebacterium. While the percentage of Parabacteroides bacteria decreased in the control group, it increased in the OSCC group, and the opposite was observed for Corynebacterium. The metatranscriptomic analysis revealed overexpression of the same metabolic signatures associated with OSCC regardless of the community profile. These included nitrogen transport, response to stress, interspecies interactions, Wnt pathway modulation, and amino acid and lipid biosynthesis. Thus, these results seem to suggest that certain collective physiological activities are critical for microbiome-mediated OSCC progression. IMPORTANCE There is growing evidence that changes in the microbiome are associated with carcinogenesis. To date, no consistent oral microbiome composition associated with OSCC has been identified. Longitudinal and functional studies like the study presented here should yield a better understanding of the role that the oral microbiome plays in OSCC. Our findings, obtained using a germ-free mouse model, indicate that the presence of different oral microbiomes enhances tumorigenesis and increases the final number of tumors in mice. By studying community-wide expression profiles, we found that regardless of the phylogenetic composition of the microbiome, the same metabolic activities were consistently associated with OSCC. Therefore, due to the functional redundancy of the microbiome, the critical element in explaining the contribution of the microbiota in OSCC is the collective physiological activity of the community, thus accounting for the previous inability to identify a consensus community profile or etiologic agents for OSCC. American Society for Microbiology 2019-08-06 /pmc/articles/PMC6687944/ /pubmed/31387932 http://dx.doi.org/10.1128/mSystems.00323-19 Text en Copyright © 2019 Stashenko et al. https://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Research Article
Stashenko, Philip
Yost, Susan
Choi, Yoonhee
Danciu, Theodora
Chen, Tsute
Yoganathan, Subbiah
Kressirer, Christine
Ruiz-Tourrella, Montserrat
Das, Bikul
Kokaras, Alexis
Frias-Lopez, Jorge
The Oral Mouse Microbiome Promotes Tumorigenesis in Oral Squamous Cell Carcinoma
title The Oral Mouse Microbiome Promotes Tumorigenesis in Oral Squamous Cell Carcinoma
title_full The Oral Mouse Microbiome Promotes Tumorigenesis in Oral Squamous Cell Carcinoma
title_fullStr The Oral Mouse Microbiome Promotes Tumorigenesis in Oral Squamous Cell Carcinoma
title_full_unstemmed The Oral Mouse Microbiome Promotes Tumorigenesis in Oral Squamous Cell Carcinoma
title_short The Oral Mouse Microbiome Promotes Tumorigenesis in Oral Squamous Cell Carcinoma
title_sort oral mouse microbiome promotes tumorigenesis in oral squamous cell carcinoma
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6687944/
https://www.ncbi.nlm.nih.gov/pubmed/31387932
http://dx.doi.org/10.1128/mSystems.00323-19
work_keys_str_mv AT stashenkophilip theoralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT yostsusan theoralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT choiyoonhee theoralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT danciutheodora theoralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT chentsute theoralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT yoganathansubbiah theoralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT kressirerchristine theoralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT ruiztourrellamontserrat theoralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT dasbikul theoralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT kokarasalexis theoralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT friaslopezjorge theoralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT stashenkophilip oralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT yostsusan oralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT choiyoonhee oralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT danciutheodora oralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT chentsute oralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT yoganathansubbiah oralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT kressirerchristine oralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT ruiztourrellamontserrat oralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT dasbikul oralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT kokarasalexis oralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma
AT friaslopezjorge oralmousemicrobiomepromotestumorigenesisinoralsquamouscellcarcinoma