1
|
Hayakawa K, Saito S, Miyoshi-Akiyama T, Fukui Y, Takemoto N, Hashimoto T, Inagaki T, Hirose K, Kobayashi K, Koizumi R, Endo M, Komatsubara M, Nomoto H, Inada M, Ide S, Kamegai K, Ashida S, Nagata N, Kato H, Ohmagari N. Comparison of the effects of cefmetazole and meropenem on microbiome: A pilot study. J Infect Chemother 2024; 30:1274-1279. [PMID: 38879077 DOI: 10.1016/j.jiac.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Cefmetazole (CMZ) is a carbapenem-sparing option in the treatment of extended-spectrum beta-lactamase (ESBL)-producing bacterial infection. In this pilot study, we aimed to compare the effects of antimicrobial treatment (meropenem [MP] and CMZ) with those of no antimicrobial treatment (control group) on the microbiome. METHODS The study was a multicenter, prospective, observational pilot study conducted from October 2020 to October 2022. Feces and saliva samples were collected for microbiome analyses at two time points (early-period: days 1-3; and late-period: days 4-30) for the antimicrobial treatment group, and at one time point for the control group. RESULTS Five feces (MP-F and CMZ-F) and five saliva (MP-S and CMZ-S) samples were included in the MP and the CMZ groups. Ten feces (C-F) and saliva (C-S) samples were included in the control group. Group α diversity was notably lower in the late-period MP-F group than the control group as determined with the Shannon richness index. β diversity analysis of the feces samples based on weighted and unweighted UniFrac distances revealed distinctions in both the late-period CMZ-F and MP-F groups compared with the control group. Weighted UniFrac analysis showed that only the early-period MP-F group differed from the control group. In the saliva samples, weighted and unweighted UniFrac analyses showed significant differences between the control group and the early CMZ, late CMZ, and late MP groups. CONCLUSIONS MP treatment may cause larger impact on the feces microbiome than CMZ in Japanese patients.
Collapse
Affiliation(s)
- Kayoko Hayakawa
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan; AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan.
| | - Sho Saito
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan; AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tohru Miyoshi-Akiyama
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Yuto Fukui
- Department of Infectious Diseases, Toho University Omori Medical Center, Tokyo, Japan
| | - Norihiko Takemoto
- Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | | | - Takeshi Inagaki
- Department of General Internal Medicine, National Center for Global Health and Medicine, Tokyo, Japan
| | - Keika Hirose
- Department of Emergency Medicine and Critical Care, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kentaro Kobayashi
- Department of Emergency Medicine and Critical Care, National Center for Global Health and Medicine, Tokyo, Japan
| | - Ryuji Koizumi
- AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mio Endo
- AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Mika Komatsubara
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hidetoshi Nomoto
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Makoto Inada
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Satoshi Ide
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Kohei Kamegai
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Shinobu Ashida
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| | - Naoyoshi Nagata
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan; Department of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hideaki Kato
- Infection Prevention and Control Department, Yokohama City University Hospital, Kanagawa, Japan
| | - Norio Ohmagari
- Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan; AMR Clinical Reference Center, Disease Control and Prevention Center, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
2
|
Simunic M, McGraw K, Pavletic SZ, Rashidi A. Intestinal microbiome and myelodysplastic syndromes: Current state of knowledge and perspectives for future. Semin Hematol 2024; 61:442-448. [PMID: 39551677 PMCID: PMC11646173 DOI: 10.1053/j.seminhematol.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 11/19/2024]
Abstract
The intestinal microbiome has been mechanistically linked with health and many disease processes. Cancer is no exception. Both in solid tumors and hematologic malignancies, there is increasing evidence supporting the involvement of the intestinal microbiome in tumor development, disease progression, response to treatment, and treatment toxicity. Consistent with microbiome mediation of the immune system and the potent effect of the immune system on cancer, the most compelling evidence has been obtained in the setting of cancer immunotherapy. Here, we review the current state of knowledge about microbiome effects in myelodysplastic syndromes, identify gaps and challenges in related research, and provide insights for future work.
Collapse
Affiliation(s)
- Marin Simunic
- Immune Deficiency Cellular Therapy Program (ID-CTP), National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Myeloid Malignancies Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Department of Hematology, Clinic for Internal Medicine, Clinical Hospital Center, Split, Croatia
| | - Kathy McGraw
- Immune Deficiency Cellular Therapy Program (ID-CTP), National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Myeloid Malignancies Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Steven Z Pavletic
- Immune Deficiency Cellular Therapy Program (ID-CTP), National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Myeloid Malignancies Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Armin Rashidi
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
3
|
Ryu EP, Gautam Y, Proctor DM, Bhandari D, Tandukar S, Gupta M, Gautam GP, Relman DA, Shibl AA, Sherchand JB, Jha AR, Davenport ER. Nepali oral microbiomes reflect a gradient of lifestyles from traditional to industrialized. MICROBIOME 2024; 12:228. [PMID: 39497165 PMCID: PMC11533410 DOI: 10.1186/s40168-024-01941-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/27/2024] [Indexed: 11/06/2024]
Abstract
BACKGROUND Lifestyle plays an important role in shaping the gut microbiome. However, its contributions to the oral microbiome remain less clear, due to the confounding effects of geography and methodology in investigations of populations studied to date. Furthermore, while the oral microbiome seems to differ between foraging and industrialized populations, we lack insight into whether transitions to and away from agrarian lifestyles shape the oral microbiota. Given the growing interest in so-called "vanishing microbiomes" potentially being a risk factor for increased disease prevalence in industrialized populations, it is important that we distinguish lifestyle from geography in the study of microbiomes across populations. RESULTS Here, we investigate salivary microbiomes of 63 Nepali individuals representing a spectrum of lifestyles: foraging, subsistence farming (individuals that transitioned from foraging to farming within the last 50 years), agriculturalists (individuals that have transitioned to farming for at least 300 years), and industrialists (expatriates that immigrated to the USA within the last 20 years). We characterize the role of lifestyle in microbial diversity, identify microbes that differ between lifestyles, and pinpoint specific lifestyle factors that may be contributing to differences in the microbiomes across populations. Contrary to prevailing views, when geography is controlled for, oral microbiome alpha diversity does not differ significantly across lifestyles. Microbiome composition, however, follows the gradient of lifestyles from foraging through agrarianism to industrialism, supporting the notion that lifestyle indeed plays a role in the oral microbiome. Relative abundances of several individual taxa, including Streptobacillus and an unclassified Porphyromonadaceae genus, also mirror lifestyle. Finally, we identify specific lifestyle factors associated with microbiome composition across the gradient of lifestyles, including smoking and grain sources. CONCLUSION Our findings demonstrate that by studying populations within Nepal, we can isolate an important role of lifestyle in determining oral microbiome composition. In doing so, we highlight the potential contributions of several lifestyle factors, underlining the importance of carefully examining the oral microbiome across lifestyles to improve our understanding of global microbiomes. Video Abstract.
Collapse
Affiliation(s)
- Erica P Ryu
- Department of Biology, Pennsylvania State University, University Park, PA, USA
| | - Yoshina Gautam
- Genetic Heritage Group, Program in Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Diana M Proctor
- Department of Microbiology and Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dinesh Bhandari
- Public Health Research Laboratory, Institute of Medicine, Maharajgunj, Kathmandu, Nepal
- School of Public Health, University of Adelaide, Adelaide, SA, Australia
| | - Sarmila Tandukar
- Public Health Research Laboratory, Institute of Medicine, Maharajgunj, Kathmandu, Nepal
- Organization for Public Health and Environment Management, Lalitpur, Bagmati, Nepal
| | - Meera Gupta
- Department of Biology, Pennsylvania State University, University Park, PA, USA
- Sidney Kimmel Medical College, Philadelphia, PA, UAE
| | | | - David A Relman
- Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
- Section of Infectious Diseases, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Ahmed A Shibl
- Genetic Heritage Group, Program in Biology, New York University Abu Dhabi, Abu Dhabi, UAE
- Center for Genomics and Systems Biology, and Public Health Research Center, New York University Abu Dhabi, Abu Dhabi, UAE
| | | | - Aashish R Jha
- Genetic Heritage Group, Program in Biology, New York University Abu Dhabi, Abu Dhabi, UAE.
- Center for Genomics and Systems Biology, and Public Health Research Center, New York University Abu Dhabi, Abu Dhabi, UAE.
| | - Emily R Davenport
- Department of Biology, Pennsylvania State University, University Park, PA, USA.
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA.
| |
Collapse
|
4
|
Mann AE, Aumend C, Crull S, O'Connell LM, Osagie E, Akhigbe P, Obuekwe O, Omoigberale A, Rowe M, Blouin T, Soule A, Kelly C, Burne RA, Coker MO, Richards VP. HIV Infection and Exposure Increases Cariogenic Taxa, Reduces Taxonomic Turnover, and Homogenizes Spatial Differentiation for the Supragingival Microbiome. RESEARCH SQUARE 2024:rs.3.rs-4720457. [PMID: 39149457 PMCID: PMC11326420 DOI: 10.21203/rs.3.rs-4720457/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Background The oral microbiome comprises distinct microbial communities that colonize diverse ecological niches across the oral cavity, the composition of which are influenced by nutrient and substrate availability, host genetics, diet, behavior, age, and other diverse host and environmental factors. Unlike other densely populated human-associated microbial ecosystems (e.g., gut, urogenital), the oral microbiome is regularly and directly exposed to the external environment and is therefore likely less stable over time. Cross sectional studies of the oral microbiome capture a glimpse of this temporal dynamism, yet a full appreciation of the relative stability, robusticity, and spatial structure of the oral environment is necessary to understand the role of microbial communities in promoting health or disease. Results Here we investigate the spatial and temporal stability of the oral microbiome over three sampling time points in the context of HIV infection and exposure. Individual teeth were sampled from a cohort of 565 Nigerian children with varying levels of tooth decay severity (i.e., caries disease). We collected 1,960 supragingival plaque samples and characterized the oral microbiome using a metataxonomic approach targeting an approximately 478 bp region of the bacterial rpoC gene. We found that both infection and exposure to HIV have significant effects on the stability of the supragingival plaque microbiome at both the spatial and temporal scale. Specifically, we detect (1) significantly lower taxonomic turnover of the oral community among exposed and infected children compared to unexposed children, (2) we find that HIV infection homogenizes the oral community across the anterior and posterior dentition, and (3) that impaired immunity (i.e., low CD4 count) and low taxonomic turnover over time in children living with HIV is associated with higher frequency of cariogenic taxa including Streptococcus mutans. Conclusions Our results document substantial community fluctuations over time in children unexposed to HIV independent of oral health status. This suggests that the oral community, under typical conditions, rapidly adapts to environmental perturbations to maintain homeostasis and that long-term taxonomic rigidity is a signal of community dysfunction, potentially leading to a higher incidence of oral disease including caries.
Collapse
|
5
|
Ryu EP, Gautam Y, Proctor DM, Bhandari D, Tandukar S, Gupta M, Gautam GP, Relman DA, Shibl AA, Sherchand JB, Jha AR, Davenport ER. Nepali oral microbiomes reflect a gradient of lifestyles from traditional to industrialized. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601557. [PMID: 39005279 PMCID: PMC11244963 DOI: 10.1101/2024.07.01.601557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Background Lifestyle plays an important role in shaping the gut microbiome. However, its contributions to the oral microbiome remains less clear, due to the confounding effects of geography and methodology in investigations of populations studied to date. Furthermore, while the oral microbiome seems to differ between foraging and industrialized populations, we lack insight into whether transitions to and away from agrarian lifestyles shape the oral microbiota. Given the growing interest in so-called 'vanishing microbiomes' potentially being a risk factor for increased disease prevalence in industrialized populations, it is important that we distinguish lifestyle from geography in the study of microbiomes across populations. Results Here, we investigate salivary microbiomes of 63 Nepali individuals representing a spectrum of lifestyles: foraging, subsistence farming (individuals that transitioned from foraging to farming within the last 50 years), agriculturalists (individuals that have transitioned to farming for at least 300 years), and industrialists (expatriates that immigrated to the United States within the last 20 years). We characterize the role of lifestyle in microbial diversity, identify microbes that differ between lifestyles, and pinpoint specific lifestyle factors that may be contributing to differences in the microbiomes across populations. Contrary to prevailing views, when geography is controlled for, oral microbiome alpha diversity does not differ significantly across lifestyles. Microbiome composition, however, follows the gradient of lifestyles from foraging through agrarianism to industrialism, supporting the notion that lifestyle indeed plays a role in the oral microbiome. Relative abundances of several individual taxa, including Streptobacillus and an unclassified Porphyromonadaceae genus, also mirror lifestyle. Finally, we identify specific lifestyle factors associated with microbiome composition across the gradient of lifestyles, including smoking and grain source. Conclusion Our findings demonstrate that by controlling for geography, we can isolate an important role for lifestyle in determining oral microbiome composition. In doing so, we highlight the potential contributions of several lifestyle factors, underlining the importance of carefully examining the oral microbiome across lifestyles to improve our understanding of global microbiomes.
Collapse
Affiliation(s)
- Erica P. Ryu
- Department of Biology, Pennsylvania State University, University Park, PA
| | - Yoshina Gautam
- Genetic Heritage Group, Program in Biology, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Diana M. Proctor
- Microbial Genomics Section, Translational and Functional Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Dinesh Bhandari
- Public Health Research Laboratory, Institute of Medicine, Maharajgunj, Kathmandu, Nepal
- School of Public Health, University of Adelaide, South Australia, Australia
| | - Sarmila Tandukar
- Public Health Research Laboratory, Institute of Medicine, Maharajgunj, Kathmandu, Nepal
- Organization for Public Health and Environment Management, Lalitpur, Bagmati, Nepal
| | - Meera Gupta
- Department of Biology, Pennsylvania State University, University Park, PA
| | | | - David A. Relman
- Departments of Medicine, and of Microbiology & Immunology, Stanford University, Stanford, CA
- Section of Infectious Diseases, Veterans Affairs Palo Alto Health Care System, Palo Alto, CA
| | - Ahmed A. Shibl
- Genetic Heritage Group, Program in Biology, New York University Abu Dhabi, Abu Dhabi, UAE
- Center for Genomics and Systems Biology, and Public Health Research Center, New York University Abu Dhabi, Abu Dhabi, UAE
| | | | - Aashish R. Jha
- Genetic Heritage Group, Program in Biology, New York University Abu Dhabi, Abu Dhabi, UAE
- Center for Genomics and Systems Biology, and Public Health Research Center, New York University Abu Dhabi, Abu Dhabi, UAE
| | - Emily R. Davenport
- Department of Biology, Pennsylvania State University, University Park, PA
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA
| |
Collapse
|
6
|
Luo C, Zhang W, Zhu J, Qiu T, Fang Q. Interleukin-2 mediated associations between gut microbiota and acute myeloid leukemia: A population-based mediation Mendelian randomization study. Heliyon 2024; 10:e33194. [PMID: 39022041 PMCID: PMC11252755 DOI: 10.1016/j.heliyon.2024.e33194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 07/20/2024] Open
Abstract
The relationship between the gut microbiota and acute myeloid leukemia (AML) has been established, but the exact role of interleukin (IL) in mediating this relationship has remained unclear. This study aimed to utilize whether interleukins mediate the relationships between gut microbiota and AML, thereby identifying potential novel targets for future AML treatment. Mendelian randomization (MR) is a method for finding the causality of exposure and outcome. Final instrumental variables were selected based on MR assumptions, and used to judge validity of the results. Our study identified risk and protective factors for AML, and interleukin-related gut microbiota. Finally, mediation MR analyses resulted in Interleukin-2 (IL-2) mediated associations between Clostridiaceae 1, Clostridium sensu stricto 1 and AML, with IL-2 respectively explaining 13.96 % and 12.11 % of the total effect of the aforementioned gut microbiota on AML. Our results successfully identified causal effects between specific gut microbiota, AML, and interleukins, while also elucidating the mediating role of IL-2 in these associations using MR analysis. These findings provide valuable insights into potential therapeutic targets for AML treatment.
Collapse
Affiliation(s)
- Chenxi Luo
- School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Wei Zhang
- School of Nursing, Wuhan University, Wuhan, 430071, China
| | - Jicheng Zhu
- School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Tianlai Qiu
- School of Nursing, Wuhan University, Wuhan, 430071, China
| | - Qingbo Fang
- School of Nursing, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
7
|
Furci F, Cicero N, Allegra A, Gangemi S. Microbiota, Diet and Acute Leukaemia: Tips and Tricks on Their Possible Connections. Nutrients 2023; 15:4253. [PMID: 37836537 PMCID: PMC10574113 DOI: 10.3390/nu15194253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/15/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
Acute leukaemia is probably one of the most recurrent cancers in children and younger adults, with an incidence of acute lymphoblastic leukaemia in 80% of cases and an incidence of acute myeloid leukaemia in 15% of cases. Yet, while incidence is common in children and adolescents, acute leukaemia is a rare disease whose aetiology still requires further analysis. Many studies have investigated the aetiology of acute leukaemia, reporting that the formation of gut microbiota may be modified by the start and development of many diseases. Considering that in patients affected by acute lymphoblastic leukaemia, there is an inherent disequilibrium in the gut microbiota before treatment compared with healthy patients, increasing evidence shows how dysbiosis of the gut microbiota provokes an inflammatory immune response, contributing to the development of cancer. Our analysis suggeststhe key role of gut microbiota in the modulation of the efficacy of leukaemia treatment as well as in the progress of many cancers, such as acute leukaemia. Therefore, in this paper, we present an examination of information found in literature regarding the role of dietary factors and gut microbiota alterations in the development of leukaemia and suggest possible future preventive and therapeutic strategies.
Collapse
Affiliation(s)
- Fabiana Furci
- Provincial Healthcare Unit, Section of Allergy, 89900 Vibo Valentia, Italy;
| | - Nicola Cicero
- Department of Biomedical, Dental, Morphological and Functional Imaging Sciences, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Allergy and Clinical Immunology Unit, Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| |
Collapse
|
8
|
Amieva-Balmori M, García-Mazcorro JF, Martínez-Conejo A, Hernández-Ramírez GA, García-Zermeño KR, Rodríguez-Aguilera O, Aja-Cadena M, Barradas-Cortés M, Quigley EMM, Remes-Troche JM. Fecal bacterial microbiota in constipated patients before and after eight weeks of daily Bifidobacterium infantis 35624 administration. REVISTA DE GASTROENTEROLOGIA DE MEXICO (ENGLISH) 2023; 88:369-380. [PMID: 35810091 DOI: 10.1016/j.rgmxen.2022.06.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 10/17/2022]
Abstract
INTRODUCTION AND AIM In recent years, probiotics have been used in functional gastrointestinal disorders, including chronic constipation (CC). The effect of Bifidobacterium infantis strain 35624 on the gut microbiota of CC patients has not been previously studied. Our aim was to analyze the fecal microbiota of constipated patients, before and after consuming a single-strain probiotic (B. infantis strain 35624). MATERIALS AND METHODS We used 16S rRNA gene high-throughput sequencing to analyze the fecal microbiota of female patients (n=13) with CC. Patients were instructed to ingest one capsule of Alflorex® (containing 1×109 CFUs/g B. infantis strain 35624) daily for eight weeks. Fecal samples were obtained at the baseline and end (final) of probiotic administration. RESULTS Alpha diversity metrics did not differ between the baseline and final periods. The butyrate producer, Oscillospira, was the taxon most strongly correlated with amplicon sequence variants (R2=0.55, p<0.0001). Except for a few bacterial taxa, there were no significant differences in relative abundance between the baseline and final periods. Beta-diversity measures also showed limited evidence for the differences between the two time periods. CONCLUSIONS The results suggest that the fecal bacterial microbiota remains stable in constipated women consuming a single-strain probiotic. Those findings may be helpful in better understanding probiotic functioning in patients with digestive disorders.
Collapse
Affiliation(s)
- M Amieva-Balmori
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - J F García-Mazcorro
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - A Martínez-Conejo
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - G A Hernández-Ramírez
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - K R García-Zermeño
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - O Rodríguez-Aguilera
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - M Aja-Cadena
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - M Barradas-Cortés
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México
| | - E M M Quigley
- Lynda K and David M Underwood Center for Digestive Disorders, Houston Methodist Hospital and Weill Cornell Medical College, Houston, TX, USA
| | - J M Remes-Troche
- Laboratorio de Fisiología Digestiva y Motilidad Gastrointestinal, Instituto de Investigaciones Medico-Biológicas, Universidad Veracruzana, Veracruz, México.
| |
Collapse
|
9
|
Wang B, Hu W, Zhang X, Cao Y, Shao L, Xu X, Liu P. Sarcopenia and gut microbiota alterations in patients with hematological diseases before and after hematopoietic stem cell transplantation. Chin J Cancer Res 2023; 35:386-398. [PMID: 37691890 PMCID: PMC10485920 DOI: 10.21147/j.issn.1000-9604.2023.04.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/20/2023] [Indexed: 09/12/2023] Open
Abstract
Objective The aim of this study was to investigate the prevalence of sarcopenia (SP) and its relationship with gut microbiota alterations in patients with hematological diseases before and after hematopoietic stem cell transplantation (HSCT). Methods A total of 108 patients with various hematological disorders were selected from Peking University People's Hospital. SP was screened and diagnosed based on the 2019 Asian Sarcopenia Diagnosis Strategy. Physical measurements and fecal samples were collected, and 16S rRNA gene sequencing was conducted. Alpha and beta diversity analyses were performed to evaluate gut microbiota composition and diversity. Results After HSCT, significant decreases in calf circumference and body mass index (BMI) were observed, accompanied by a decline in physical function. Gut microbiota analyses revealed significant differences in the relative abundance of Enterococcus, Bacteroides, Blautia and Dorea species before and after HSCT (P<0.05). Before HSCT, sarcopenic patients had lower Dorea levels and higher Phascolarctobacterium levels than non-sarcopenia patients (P<0.01). After HSCT, no significant differences in species abundance were observed. Alpha diversity analysis showed significant differences in species diversity among the groups, with the highest diversity in the post-HSCT 90-day group and the lowest in the post-HSCT 30-day group. Beta diversity analysis revealed significant differences in species composition between pre- and post-HSCT time points but not between SP groups. Linear discriminant analysis effect size (LEfSe) identified Alistipes, Rikenellaceae, Alistipes putredinis, Prevotellaceae defectiva and Blautia coccoides as biomarkers for the pre-HSCT sarcopenia group. Functional predictions showed significant differences in anaerobic, biofilm-forming and oxidative stress-tolerant functions among the groups (P<0.05). Conclusions This study demonstrated a significant decline in physical function after HSCT and identified potential gut microbiota biomarkers and functional alterations associated with SP in patients with hematological disorders. Further research is needed to explore the underlying mechanisms and potential therapeutic targets.
Collapse
Affiliation(s)
- Boshi Wang
- Department of Clinical Nutrition, Peking University People’s Hospital, Beijing 100044, China
| | - Wei Hu
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| | - Xue Zhang
- Department of Clinical Nutrition, Peking University People’s Hospital, Beijing 100044, China
| | - Yanchao Cao
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| | - Lin Shao
- Department of Clinical Nutrition, Peking University People’s Hospital, Beijing 100044, China
| | - Xiaodong Xu
- Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing 100044, China
| | - Peng Liu
- Department of Clinical Nutrition, Peking University People’s Hospital, Beijing 100044, China
| |
Collapse
|
10
|
Gavin NC, Larsen E, Runnegar N, Mihala G, Keogh S, McMillan D, Ray‐Barruel G, Rickard CM. Association between parenteral nutrition-containing intravenous lipid emulsion and bloodstream infections in patients with single-lumen central venous access: A secondary analysis of a randomized trial. JPEN J Parenter Enteral Nutr 2023; 47:783-795. [PMID: 37288612 PMCID: PMC10946626 DOI: 10.1002/jpen.2530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 05/12/2023] [Accepted: 06/06/2023] [Indexed: 06/09/2023]
Abstract
BACKGROUND Distinguishing primary bloodstream infections (BSIs) related to central venous access devices (CVADs) from those that occur through other mechanisms, such as a damaged mucosal barrier, is difficult. METHODS Secondary analysis was conducted on data from patients with CVADs that were collected for a large, randomized trial. Patients were divided into two groups: those who received parenteral nutrition (PN)-containing intravenous lipid emulsion (ILE) and those who did not have PN-containing ILE. This study investigated the influence of PN-containing ILE (ILE PN) on primary BSIs in patients with a CVAD. RESULTS Of the 807 patients, 180 (22%) received ILE PN. Most (627/807; 73%) were recruited from the hematology and hematopoietic stem cell transplant unit, followed by surgical (90/807; 11%), trauma and burns (61/807; 8%), medical (44/807; 5%), and oncology (23/807; 3%). When primary BSI was differentiated as a central line-associated BSI (CLABSI) or mucosal barrier injury laboratory-confirmed BSI (MBI-LCBI), the incidence of CLABSI was similar in the ILE PN and non-ILE PN groups (15/180 [8%] vs 57/627 [9%]; P = 0.88) and the incidence of MBI-LCBI was significantly different between groups (31/180 [17%] ILE PN vs 41/627 [7%] non-ILE PN; P < 0.01). CONCLUSION Our data indicate that twice as many primary BSIs in ILE PN patients are due to MBIs than CVADs. It is important to consider the MBI-LCBI classification, as some CLABSI prevention efforts aimed at CVADs for the ILE PN population may be better directed to gastrointestinal tract protection interventions.
Collapse
Affiliation(s)
- Nicole C. Gavin
- Cancer Care ServicesRoyal Brisbane and Women's HospitalQueenslandAustralia
- Alliance for Vascular Access Teaching and Research Group, School of Nursing and MidwiferyGriffith UniversityQueenslandAustralia
- Centre for Healthcare TransformationQueensland University of TechnologyQueenslandAustralia
- School of NursingQueensland University of TechnologyQueenslandAustralia
- School of Nursing, Midwifery and Social WorkThe University of QueenslandSt LuciaQueenslandAustralia
- Faculty of MedicineUniversity of QueenslandQueenslandAustralia
| | - Emily Larsen
- Alliance for Vascular Access Teaching and Research Group, School of Nursing and MidwiferyGriffith UniversityQueenslandAustralia
- Nursing and Midwifery Research CentreRoyal Brisbane and Women's HospitalHerstonAustralia
- Patient‐Centre Health Services, Menzies Health Institute QueenslandGriffith UniversityQueenslandAustralia
| | - Naomi Runnegar
- Faculty of MedicineUniversity of QueenslandQueenslandAustralia
- Infection Management ServicesPrincess Alexandra HospitalQueenslandAustralia
| | - Gabor Mihala
- Alliance for Vascular Access Teaching and Research Group, School of Nursing and MidwiferyGriffith UniversityQueenslandAustralia
- School of Medicine and DentistryGriffith UniversityQueenslandAustralia
| | - Samantha Keogh
- Alliance for Vascular Access Teaching and Research Group, School of Nursing and MidwiferyGriffith UniversityQueenslandAustralia
- Centre for Healthcare TransformationQueensland University of TechnologyQueenslandAustralia
- School of NursingQueensland University of TechnologyQueenslandAustralia
- Nursing and Midwifery Research CentreRoyal Brisbane and Women's HospitalHerstonAustralia
| | - David McMillan
- Alliance for Vascular Access Teaching and Research Group, School of Nursing and MidwiferyGriffith UniversityQueenslandAustralia
- School of Science, Technology, Engineering and Education; Centre for BioInnovationUniversity of the Sunshine CoastQueenslandAustralia
| | - Gillian Ray‐Barruel
- Alliance for Vascular Access Teaching and Research Group, School of Nursing and MidwiferyGriffith UniversityQueenslandAustralia
- School of Nursing, Midwifery and Social WorkThe University of QueenslandSt LuciaQueenslandAustralia
- Nursing and Midwifery Research CentreRoyal Brisbane and Women's HospitalHerstonAustralia
| | - Claire M. Rickard
- Alliance for Vascular Access Teaching and Research Group, School of Nursing and MidwiferyGriffith UniversityQueenslandAustralia
- School of Nursing, Midwifery and Social WorkThe University of QueenslandSt LuciaQueenslandAustralia
- Nursing and Midwifery Research CentreRoyal Brisbane and Women's HospitalHerstonAustralia
- Herston Infectious Diseases InstituteMetro North HealthHerstonAustralia
| |
Collapse
|
11
|
McMahon S, Sahasrabhojane P, Kim J, Franklin S, Chang CC, Jenq RR, Hillhouse AE, Shelburne SA, Galloway-Peña J. Contribution of the Oral and Gastrointestinal Microbiomes to Bloodstream Infections in Leukemia Patients. Microbiol Spectr 2023; 11:e0041523. [PMID: 37022173 PMCID: PMC10269818 DOI: 10.1128/spectrum.00415-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/20/2023] [Indexed: 04/07/2023] Open
Abstract
Bloodstream infections (BSIs) pose a significant mortality risk for acute myeloid leukemia (AML) patients. It has been previously reported that intestinal domination (>30% relative abundance [RA] attributed to a single taxon) with the infecting taxa often precedes BSI in stem cell transplant patients. Using 16S rRNA amplicon sequencing, we analyzed oral and stool samples from 63 AML patients with BSIs to determine the correlation between the infectious agent and microbiome composition. Whole-genome sequencing and antimicrobial susceptibilities were performed on all BSI isolates. Species-level detection of the infectious agent and presence of antibiotic resistance determinants in the stool (blaCTX-M-15, blaCTX-M-14, cfrA, and vanA) were confirmed via digital droplet PCR (ddPCR). Individuals with Escherichia coli (stool P < 0.001), Pseudomonas aeruginosa (oral P = 0.004, stool P < 0.001), and viridans group streptococci (VGS) (oral P = 0.001) bacteremia had a significantly higher relative abundance of those respective genera than other BSI patients, which appeared to be site specific. Although 78% of patients showed presence of the infectious genera in the stool and/or saliva, only 7 exhibited microbiome domination. ddPCR confirmed species specificity of the 16S data and detected the antibiotic resistance determinants found in the BSI isolates within concurrent stools. Although gastrointestinal (GI) domination by an infecting organism was not present at the time of most BSIs in AML, the pathogens, along with AMR elements, were detectable in the majority of patients. Thus, rapid genetic assessment of oral and stool samples for the presence of potential pathogens and AMR determinants might inform personalized therapeutic approaches in immunocompromised patients with suspected infection. IMPORTANCE A major cause of mortality in hematologic malignancy patients is BSI. Previous studies have demonstrated that bacterial translocation from the GI microbiome is a major source of BSIs and is often preceded by increased levels of the infectious taxa in the GI (>30% abundance by 16S rRNA sequencing). In this study, we sought to better understand how domination and abundance levels of the oral and gut microbiome relate to bacteremia occurrence in acute myeloid leukemia patients. We conclude that analyses of both oral and stool samples can help identify BSI and antimicrobial resistance determinants, thus potentially improving the timing and tailoring of antibiotic treatment strategies for high-risk patients.
Collapse
Affiliation(s)
- Stephanie McMahon
- Interdisciplinary Genetics Program, Texas A&M University, College Station, Texas, USA
| | - Pranoti Sahasrabhojane
- Department of Infectious Diseases, Infection Control, and Employee Health, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jiwoong Kim
- Department of Bioinformatics and Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Samantha Franklin
- Interdisciplinary Genetics Program, Texas A&M University, College Station, Texas, USA
| | - Chia-Chi Chang
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Robert R. Jenq
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Andrew E. Hillhouse
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, USA
- Texas A&M Institute for Genome Sciences & Society, Texas A&M University, College Station, Texas, USA
| | - Samuel A. Shelburne
- Department of Infectious Diseases, Infection Control, and Employee Health, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jessica Galloway-Peña
- Interdisciplinary Genetics Program, Texas A&M University, College Station, Texas, USA
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Veterinary Pathobiology, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
12
|
Dora D, Ligeti B, Kovacs T, Revisnyei P, Galffy G, Dulka E, Krizsán D, Kalcsevszki R, Megyesfalvi Z, Dome B, Weiss GJ, Lohinai Z. Non-small cell lung cancer patients treated with Anti-PD1 immunotherapy show distinct microbial signatures and metabolic pathways according to progression-free survival and PD-L1 status. Oncoimmunology 2023; 12:2204746. [PMID: 37197440 PMCID: PMC10184596 DOI: 10.1080/2162402x.2023.2204746] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/13/2023] [Accepted: 04/16/2023] [Indexed: 05/19/2023] Open
Abstract
Due to the high variance in response rates concerning anti-PD1 immunotherapy (IT), there is an unmet need to discover innovative biomarkers to predict immune checkpoint inhibitor (ICI)-efficacy. Our study included 62 Caucasian advanced-stage non-small cell lung cancer (NSCLC) patients treated with anti-PD1 ICI. Gut bacterial signatures were evaluated by metagenomic sequencing and correlated with progression-free survival (PFS), PD-L1 expression and other clinicopathological parameters. We confirmed the predictive role of PFS-related key bacteria with multivariate statistical models (Lasso- and Cox-regression) and validated on an additional patient cohort (n = 60). We find that alpha-diversity showed no significant difference in any comparison. However, there was a significant difference in beta-diversity between patients with long- (>6 months) vs. short (≤6 months) PFS and between chemotherapy (CHT)-treated vs. CHT-naive cases. Short PFS was associated with increased abundance of Firmicutes (F) and Actinobacteria phyla, whereas elevated abundance of Euryarchaeota was specific for low PD-L1 expression. F/Bacteroides (F/B) ratio was significantly increased in patients with short PFS. Multivariate analysis revealed an association between Alistipes shahii, Alistipes finegoldii, Barnesiella visceriola, and long PFS. In contrast, Streptococcus salivarius, Streptococcus vestibularis, and Bifidobacterium breve were associated with short PFS. Using Random Forest machine learning approach, we find that taxonomic profiles performed superiorly in predicting PFS (AUC = 0.74), while metabolic pathways including Amino Acid Synthesis and Fermentation were better predictors of PD-L1 expression (AUC = 0.87). We conclude that specific metagenomic features of the gut microbiome, including bacterial taxonomy and metabolic pathways might be suggestive of ICI efficacy and PD-L1 expression in NSCLC patients.
Collapse
Affiliation(s)
- David Dora
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
| | - Balazs Ligeti
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Tamas Kovacs
- Department of Anatomy, Histology and Embryology, Semmelweis University, Budapest, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Peter Revisnyei
- Department of Telecommunications and Media Informatics, Budapest University of Technology and Economics, Budapest, Hungary
| | | | - Edit Dulka
- County Hospital of Torokbalint, Torokbalint, Hungary
| | - Dániel Krizsán
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Regina Kalcsevszki
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Zsolt Megyesfalvi
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery, National Institute of Oncology, Semmelweis University, Budapest, Hungary
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Balazs Dome
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery, National Institute of Oncology, Semmelweis University, Budapest, Hungary
- Department of Thoracic Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Department of Translational Medicine, Lund University, Sweden
| | - Glen J. Weiss
- UMass Chan Medical School, Department of Medicine, Worcester, MA, USA
| | - Zoltan Lohinai
- County Hospital of Torokbalint, Torokbalint, Hungary
- Translational Medicine Institute, Semmelweis University, Budapest, Hungary
| |
Collapse
|
13
|
Takaori A, Hashimoto D, Ikeura T, Ito T, Nakamaru K, Masuda M, Nakayama S, Yamaki S, Yamamoto T, Fujimoto K, Matsuo Y, Akagawa S, Ishida M, Yamaguchi K, Imoto S, Hirota K, Uematsu S, Satoi S, Sekimoto M, Naganuma M. Impact of neoadjuvant therapy on gut microbiome in patients with resectable/borderline resectable pancreatic ductal adenocarcinoma. Pancreatology 2023:S1424-3903(23)00074-1. [PMID: 37088586 DOI: 10.1016/j.pan.2023.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/22/2023] [Accepted: 04/03/2023] [Indexed: 04/25/2023]
Abstract
BACKGROUND /Objectives: Effects of chemotherapy on gut microbiota have been reported in various carcinomas. The current study aimed to evaluate the changes in the gut microbiota before and after neoadjuvant chemotherapy (NAC) in patients with resectable (R) and borderline resectable (BR) pancreatic ductal adenocarcinoma (PDAC) and understand their clinical implications. METHODS Twenty patients diagnosed with R/BR-PDAC were included in this study. Stool samples were collected at two points, before and after NAC, for microbiota analysis using 16S ribosomal RNA (16S rRNA) gene sequences. RESULTS Of the 20 patients, 18 (90%) were treated with gemcitabine plus S-1 as NAC, and the remaining patients received gemcitabine plus nab-paclitaxel and a fluorouracil, leucovorin, irinotecan, and oxaliplatin combination. No significant differences were observed in the α- and β-diversity before and after NAC. Bacterial diversity was not associated with Evans classification (histological grade of tumor destruction by NAC) or postoperative complications. The relative abundance of Actinobacteria phylum after NAC was significantly lower than that before NAC (P = 0.02). At the genus level, the relative abundance of Bifidobacterium before NAC in patients with Evans grade 2 disease was significantly higher than that in patients with Evans grade 1 disease (P = 0.03). Patients with Evans grade 2 lost significantly more Bifidobacterium than patients with Evans grade 1 (P = 0.01). CONCLUSIONS The diversity of gut microbiota was neither decreased by NAC for R/BR-PDAC nor associated with postoperative complications. Lower incidence of Bifidobacterium genus before NAC may be associated with a lower pathological response to NAC.
Collapse
Affiliation(s)
- Ayaka Takaori
- Third Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | | | - Tsukasa Ikeura
- Third Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Takashi Ito
- Third Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Koh Nakamaru
- Third Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Masataka Masuda
- Third Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Shinji Nakayama
- Third Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - So Yamaki
- Department of Surgery, Kansai Medical University, Osaka, Japan
| | | | - Kosuke Fujimoto
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan; Division of Metagenome Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yoshiyuki Matsuo
- Department of Human Stress Response Science, Kansai Medical University, Osaka, Japan
| | - Shohei Akagawa
- Department of Pediatrics, Kansai Medical University, Osaka, Japan
| | - Mitsuaki Ishida
- Department of Pathology, Osaka Medical and Pharmaceutical University, Osaka, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kiichi Hirota
- Department of Human Stress Response Science, Kansai Medical University, Osaka, Japan
| | - Satoshi Uematsu
- Department of Immunology and Genomics, Graduate School of Medicine, Osaka Metropolitan University, Osaka, Japan; Division of Metagenome Medicine, Human Genome Center, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sohei Satoi
- Department of Surgery, Kansai Medical University, Osaka, Japan; Division of Surgical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Makoto Naganuma
- Third Department of Internal Medicine, Kansai Medical University, Osaka, Japan.
| |
Collapse
|
14
|
Park DJ, Plantinga AM. Impact of Data and Study Characteristics on Microbiome Volatility Estimates. Genes (Basel) 2023; 14:genes14010218. [PMID: 36672959 PMCID: PMC9859452 DOI: 10.3390/genes14010218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/10/2023] [Accepted: 01/12/2023] [Indexed: 01/19/2023] Open
Abstract
The human microbiome is a dynamic community of bacteria, viruses, fungi, and other microorganisms. Both the composition of the microbiome (the microbes that are present and their relative abundances) and the temporal variability of the microbiome (the magnitude of changes in their composition across time, called volatility) has been associated with human health. However, the effect of unbalanced sampling intervals and differential read depth on the estimates of microbiome volatility has not been thoroughly assessed. Using four publicly available gut and vaginal microbiome time series, we subsampled the datasets to several sampling intervals and read depths and then compared additive, multiplicative, centered log ratio (CLR)-based, qualitative, and distance-based measures of microbiome volatility between the conditions. We find that longer sampling intervals are associated with larger quantitative measures of change (particularly for common taxa), but not with qualitative measures of change or distance-based volatility quantification. A lower sequencing read depth is associated with smaller multiplicative, CLR-based, and qualitative measures of change (particularly for less common taxa). Strategic subsampling may serve as a useful sensitivity analysis in unbalanced longitudinal studies investigating clinical associations with microbiome volatility.
Collapse
Affiliation(s)
| | - Anna M. Plantinga
- Department of Mathematics and Statistics, Williams College, Williamstown, MA 01267, USA
- Correspondence:
| |
Collapse
|
15
|
Omori M, Kato-Kogoe N, Sakaguchi S, Komori E, Inoue K, Yamamoto K, Hamada W, Hayase T, Tano T, Nakamura S, Nakano T, Une H, Ueno T. Characterization of Oral Microbiota Following Chemotherapy in Patients With Hematopoietic Malignancies. Integr Cancer Ther 2023; 22:15347354231159309. [PMID: 36922730 PMCID: PMC10021090 DOI: 10.1177/15347354231159309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Abstract
Oral microbiota may be associated with serious local or systemic medical conditions resulting from chemotherapy. This study was conducted to evaluate the changes in the oral microbiota following the initiation of chemotherapy in patients with hematopoietic malignancies and to identify the characteristics of the oral microbiota associated with oral mucositis. Oral samples were collected from 57 patients with hematopoietic malignancies at 2 time points: before the start of chemotherapy and 8 to 20 days after the start of chemotherapy, when chemotherapy-induced oral mucositis often occurs, and 16S rRNA metagenomic analyses were performed. Comparative and linear discriminant analysis effect size (LEfSe) analyses were used to determine the characteristic bacterial groups before and after the initiation of chemotherapy and in those who developed oral mucositis. The alpha and beta diversities of oral microbiota before and after the initiation of chemotherapy differed significantly (operational taxonomic unit index, P < .001; Shannon's index, P < .001; unweighted UniFrac distances, P = .001; and weighted UniFrac distances, P = .001). The LEfSe analysis revealed a group of bacteria whose abundance differed significantly before and after the initiation of chemotherapy. In the group of patients who developed oral mucositis, a characteristic group of bacteria was identified before the start of chemotherapy. In conclusion, we characterized the oral microbiota associated with the initiation of chemotherapy in patients with hematopoietic malignancies. In addition, our findings suggest that oral microbiota composition before the start of chemotherapy may be associated with oral mucositis. The results of this study emphasize the importance of oral management focusing on the oral microbiota during chemotherapy in patients with hematologic malignancies.
Collapse
Affiliation(s)
- Michi Omori
- Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | | | | | - Eri Komori
- Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Kazuya Inoue
- Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Kayoko Yamamoto
- Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Wataru Hamada
- Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Tomoyoshi Hayase
- Chugoku Central Hospital of the Mutual Aid Association of Public School Teachers, Fukuyama, Japan
| | - Tomoyuki Tano
- Chugoku Central Hospital of the Mutual Aid Association of Public School Teachers, Fukuyama, Japan
| | | | - Takashi Nakano
- Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Hidenori Une
- Chugoku Central Hospital of the Mutual Aid Association of Public School Teachers, Fukuyama, Japan
| | - Takaaki Ueno
- Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| |
Collapse
|
16
|
Zhou Y, Zhou C, Zhang A. Gut microbiota in acute leukemia: Current evidence and future directions. Front Microbiol 2022; 13:1045497. [PMID: 36532458 PMCID: PMC9751036 DOI: 10.3389/fmicb.2022.1045497] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/09/2022] [Indexed: 08/18/2023] Open
Abstract
Gut microbiota includes a large number of microorganisms inhabiting the human gastrointestinal tract, which show a wide range of physiological functions, including digestion, metabolism, immunity, neural development, etc., and are considered to play an increasingly important role in health and disease. A large number of studies have shown that gut microbiota are closely associated with the onset and development of several diseases. In particular, the interaction between gut microbiota and cancer has recently attracted scholars' attention. Acute leukemia (AL) is a common hematologic malignancy, especially in children. Microbiota can affect hematopoietic function, and the effects of chemotherapy and immunotherapy on AL are noteworthy. The composition and diversity of gut microbiota are important factors that influence and predict the complications and prognosis of AL after chemotherapy or hematopoietic stem cell transplantation. Probiotics, prebiotics, fecal microbiota transplantation, and dietary regulation may reduce side effects of leukemia therapy, improve response to treatment, and improve prognosis. This review concentrated on the role of the gut microbiota in the onset and development of AL, the response and side effects of chemotherapy drugs, infection during treatment, and therapeutic efficacy. According to the characteristics of gut microbes, the applications and prospects of microbial preparations were discussed.
Collapse
Affiliation(s)
| | | | - Aijun Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
17
|
Jiao J, Zheng Y, Zhang Q, Xia D, Zhang L, Ma N. Saliva microbiome changes in thyroid cancer and thyroid nodules patients. Front Cell Infect Microbiol 2022; 12:989188. [PMID: 36034695 PMCID: PMC9403763 DOI: 10.3389/fcimb.2022.989188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 07/21/2022] [Indexed: 11/13/2022] Open
Abstract
Objective Thyroid disease has been reported to associate with gut microbiota, but the effects of thyroid cancer and thyroid nodules on the oral microbiota are still largely unknown. This study aimed to identify the variation in salivary microbiota and their potential association with thyroid cancer and thyroid nodules. Methods We used 16S rRNA high-throughput sequencing to examine the salivary microbiota of thyroid cancer patients (n = 14), thyroid nodules patients (n = 9), and healthy controls (n = 15). Results The alpha-diversity indices Chao1 and ACE were found to be relatively higher in patients with thyroid cancer and thyroid nodules compared to healthy controls. The beta diversity in both the thyroid cancer and thyroid nodules groups was divergent from the healthy control group. The genera Alloprevotella, Anaeroglobus, Acinetobacter, unclassified Bacteroidales, and unclassified Cyanobacteriales were significantly enriched in the thyroid cancer group compared with the healthy control group. In contrast, the microbiome of the healthy controls was mainly composed of the genera Haemophilus, Lautropia, Allorhizobium Neorhizobium Pararhizobium Rhizobium, Escherichia Shigella, and unclassified Rhodobacteraceae. The thyroid nodules group was dominated by genre uncultured Candidatus Saccharibacteria bacterium, unclassified Clostridiales bacterium feline oral taxon 148, Treponema, unclassified Prevotellaceae, Mobiluncus, and Acholeplasma. In contrast, the genera unclassified Rhodobacteraceae and Aggregatibacter dominated the healthy control group. The study also found that clinical indicators were correlated with the saliva microbiome. Conclusion The salivary microbiota variation may be connected with thyroid cancer and thyroid nodules.
Collapse
Affiliation(s)
- Junjun Jiao
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Youli Zheng
- The School and Hospital of Stomatology, Tianjin Medical University, Tianjin, China
| | - Qingyu Zhang
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Degeng Xia
- Hospital of Stomatology, Jilin University, Changchun, China
| | - Li Zhang
- Hospital of Stomatology, Jilin University, Changchun, China
- *Correspondence: Ning Ma, ; Li Zhang,
| | - Ning Ma
- Hospital of Stomatology, Jilin University, Changchun, China
- *Correspondence: Ning Ma, ; Li Zhang,
| |
Collapse
|
18
|
Rashidi A, Ebadi M, Rehman TU, Elhusseini H, Halaweish H, Kaiser T, Holtan SG, Khoruts A, Weisdorf DJ, Staley C. Compilation of longitudinal gut microbiome, serum metabolome, and clinical data in acute myeloid leukemia. Sci Data 2022; 9:468. [PMID: 35918343 PMCID: PMC9346123 DOI: 10.1038/s41597-022-01600-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/27/2022] [Indexed: 12/03/2022] Open
Abstract
Induction chemotherapy for patients with acute myeloid leukemia (AML) is a unique clinical scenario. These patients spend several weeks in the hospital, receiving multiple antibiotics, experiencing gastrointestinal mucosal damage, and suffering severe impairments in their immune system and nutrition. These factors cause major disruptions to the gut microbiota to a level rarely seen in other clinical conditions. Thus, the study of the gut microbiota in these patients can reveal novel aspects of microbiota-host relationships. When combined with the circulating metabolome, such studies could shed light on gut microbiota contribution to circulating metabolites. Collectively, gut microbiota and circulating metabolome are known to regulate host physiology. We have previously deposited amplicon sequences from 566 fecal samples from 68 AML patients. Here, we provide sample-level details and a link, using de-identified patient IDs, to additional data including serum metabolomics (260 samples from 36 patients) and clinical metadata. The detailed information provided enables comprehensive multi-omics analysis. We validate the technical quality of these data through 3 examples and demonstrate a method for integrated analysis.
Collapse
Affiliation(s)
- Armin Rashidi
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA.
| | - Maryam Ebadi
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Tauseef Ur Rehman
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Heba Elhusseini
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Hossam Halaweish
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Thomas Kaiser
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
| | - Shernan G Holtan
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Daniel J Weisdorf
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | |
Collapse
|
19
|
Gao S, Khan MI, Kalsoom F, Liu Z, Chen Y, Chen Z. Role of gene regulation and inter species interaction as a key factor in gut microbiota adaptation. Arch Microbiol 2022; 204:342. [PMID: 35595857 DOI: 10.1007/s00203-022-02935-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/08/2022] [Accepted: 04/19/2022] [Indexed: 12/13/2022]
Abstract
Gut microbiota is a class of microbial flora present in various eukaryotic multicellular complex animals such as human beings. Their community's growth and survival are greatly influenced by various factors such as host-pathogen, pathogen-environment and genetic regulation. Modern technologies like metagenomics have particularly extended our capacity to uncover the microbial treasures in challenging conditions like communities surviving at high altitude. Molecular characterizations by newly developed sequencing tools have shown that this complex interaction greatly influences microbial adaptation to the environment. Literature shows that gut microbiota alters the genetic expression and switches to an alternative pathway under the influence of unfavorable conditions. The remarkable adaptability of microbial genetic regulatory networks enables them to survive and expand in tough and energy-limited conditions. Variable prevalence of species in various regions has strengthened this initial evidence. In view of the interconnection of the world in the form of a global village, this phenomenon must be explored more clearly. In this regard, recently there has been significant addition of knowledge to the field of microbial adaptation. This review summarizes and shed some light on mechanisms of microbial adaptation via gene regulation and species interaction in gut microbiota.
Collapse
Affiliation(s)
- Shuang Gao
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 260027, Anhui, People's Republic of China
| | - Muhammad Imran Khan
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 260027, Anhui, People's Republic of China. .,Department of Pathology, District Headquarters Hospital, Jhang, 35200, Punjab, Islamic Republic of Pakistan.
| | - Fadia Kalsoom
- Department of Microbiology, School of Medicine, Ajou University, Suwon, 16499, Republic of Korea
| | - Zhen Liu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Yanxin Chen
- College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China
| | - Zhengli Chen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China. .,College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, 611130, People's Republic of China.
| |
Collapse
|
20
|
Anthony WE, Wang B, Sukhum KV, D'Souza AW, Hink T, Cass C, Seiler S, Reske KA, Coon C, Dubberke ER, Burnham CAD, Dantas G, Kwon JH. Acute and persistent effects of commonly used antibiotics on the gut microbiome and resistome in healthy adults. Cell Rep 2022; 39:110649. [PMID: 35417701 PMCID: PMC9066705 DOI: 10.1016/j.celrep.2022.110649] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/07/2022] [Accepted: 03/17/2022] [Indexed: 02/09/2023] Open
Abstract
Antibiotics are deployed against bacterial pathogens, but their targeting of conserved microbial processes means they also collaterally perturb the commensal microbiome. To understand acute and persistent effects of antibiotics on the gut microbiota of healthy adult volunteers, we quantify microbiome dynamics before, during, and 6 months after exposure to 4 commonly used antibiotic regimens. We observe an acute decrease in species richness and culturable bacteria after antibiotics, with most healthy adult microbiomes returning to pre-treatment species richness after 2 months, but with an altered taxonomy, resistome, and metabolic output, as well as an increased antibiotic resistance burden. Azithromycin delays the recovery of species richness, resulting in greater compositional distance. A subset of volunteers experience a persistent reduction in microbiome diversity after antibiotics and share compositional similarities with patients hospitalized in intensive care units. These results improve our quantitative understanding of the impact of antibiotics on commensal microbiome dynamics, resilience, and recovery.
Collapse
Affiliation(s)
- Winston E Anthony
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bin Wang
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kimberley V Sukhum
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Alaric W D'Souza
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tiffany Hink
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Candice Cass
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sondra Seiler
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kimberly A Reske
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christopher Coon
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Erik R Dubberke
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Carey-Ann D Burnham
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences & Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA.
| | - Jennie H Kwon
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
21
|
Franklin S, Aitken SL, Shi Y, Sahasrabhojane PV, Robinson S, Peterson CB, Daver N, Ajami NA, Kontoyiannis DP, Shelburne SA, Galloway-Peña J. Oral and Stool Microbiome Coalescence and Its Association With Antibiotic Exposure in Acute Leukemia Patients. Front Cell Infect Microbiol 2022; 12:848580. [PMID: 35433514 PMCID: PMC9010033 DOI: 10.3389/fcimb.2022.848580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/08/2022] [Indexed: 11/16/2022] Open
Abstract
Failure to maintain segregation of oral and gut microbial communities has been linked to several diseases. We sought to characterize oral-fecal microbiome community coalescence, ectopic extension of oral bacteria, clinical variables contributing to this phenomenon, and associated infectious consequences by analyzing the 16S rRNA V4 sequences of longitudinal fecal (n=551) and oral (n=737) samples from 97 patients with acute myeloid leukemia (AML) receiving induction chemotherapy (IC). Clustering observed in permutation based multivariate analysis of variance (PERMANOVA) of Bray-Curtis dissimilarity and PCoA plot of UniFrac distances between intra-patient longitudinal oral-stool sample pairs suggested potential oral-stool microbial community coalescence. Bray-Curtis dissimilarities and UniFrac distances were used to create an objective definition of microbial community coalescence. We determined that only 23 of the 92 patients exhibited oral-stool community coalescence. This was validated through a linear mixed model which determined that patients who experienced coalescence had an increased proportion of shared to unique OTUs between their oral-stool sample pairs over time compared to non-coalesced patients. Evaluation of longitudinal microbial characteristics revealed that patients who experienced coalescence had increased stool abundance of Streptococcus and Stenotrophomonas compared to non-coalesced patients. When treated as a time-varying covariate, each additional day of linezolid (HR 1.15, 95% CI 1.06 – 1.24, P <0.001), meropenem (HR 1.13, 95% CI 1.05 – 1.21, P = 0.001), metronidazole (HR 1.13, 95% CI 1.05 – 1.21, P = 0.001), and cefepime (HR 1.10, 95% CI 1.01 – 1.18, P = 0.021) increased the hazard of oral-stool microbial community coalescence. Levofloxacin receipt was associated with a lower risk of microbiome community coalescence (HR 0.75, 95% CI 0.61 – 0.93, P = 0.009). By the time of neutrophil recovery, the relative abundance of Bacteroidia (P<0.001), Fusobacteria (P=0.012), and Clostridia (P=0.013) in the stool were significantly lower in patients with oral-gut community coalescence. Exhibiting oral-stool community coalescence was associated with the occurrence of infections prior to neutrophil recovery (P=0.002), as well as infections during the 90 days post neutrophil recovery (P=0.027). This work elucidates specific antimicrobial effects on microbial ecology and furthers the understanding of oral/intestinal microbial biogeography and its implications for adverse clinical outcomes.
Collapse
Affiliation(s)
- Samantha Franklin
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
- Interdisciplinary Graduate Program in Genetics and Genomics, Texas A&M University, College Station, TX, United States
| | - Samuel L. Aitken
- Department of Pharmacy, Michigan Medicine, Ann Arbor, MI, United States
| | - Yushi Shi
- Department of Statistics and Center for Biomedical Informatics, University of Missouri, Columbia, MO, United States
| | - Pranoti V. Sahasrabhojane
- Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sarah Robinson
- Department of Statistics, Rice University, Houston, TX, United States
| | - Christine B. Peterson
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nadim A. Ajami
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Dimitrios P. Kontoyiannis
- Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Samuel A. Shelburne
- Department of Infectious Disease, Infection Control and Employee Health, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jessica Galloway-Peña
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
- Interdisciplinary Graduate Program in Genetics and Genomics, Texas A&M University, College Station, TX, United States
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- *Correspondence: Jessica Galloway-Peña,
| |
Collapse
|
22
|
Muhammed A, D'Alessio A, Enica A, Talbot T, Fulgenzi CAM, Nteliopoulos G, Goldin RD, Cortellini A, Pinato DJ. Predictive biomarkers of response to immune checkpoint inhibitors in hepatocellular carcinoma. Expert Rev Mol Diagn 2022; 22:253-264. [PMID: 35236211 DOI: 10.1080/14737159.2022.2049244] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Hepatocellular carcinoma (HCC) is the most common primary liver cancer and fourth leading cause of cancer death. While drug discovery to improve disease survival was historically poor, there is now evidence of significant potential for immune checkpoint inhibitors (ICPIs) in treatment of the disease, and indeed such drug approvals are beginning to emerge. AREAS COVERED HCC typically arises in the context of cirrhosis and chronic liver disease (CLD), and HCC exhibits significant biological heterogeneity, in part reflecting the broad range of aetiologies of CLD. Different classes and combinations of ICPI-based therapy exist, but not all patients will respond and predictive biomarkers are not yet available to guide clinician decision making, unlike some other cancer types. In this review, we discuss the emerging biomarkers for ICPI sensitivity in HCC, including tumour genomic features, perturbation of the gut microbiome and systemic inflammatory markers. EXPERT OPINION Additional profiling studies are required to appreciate existing trends with clinical outcome and to further drive clinical studies in disease stratification by response. This will only be possible within collaborative and international efforts, especially regarding biopsy collection. A close collaboration between basic scientists and clinicians will be the key to shape the next future of HCC biomarker research.
Collapse
Affiliation(s)
| | - Antonio D'Alessio
- Department of Surgery & Cancer, Imperial College London, UK.,Department of Biomedical Sciences, Humanitas University, Italy
| | - Andrei Enica
- Department of Surgery & Cancer, Imperial College London, UK
| | - Thomas Talbot
- Department of Surgery & Cancer, Imperial College London, UK
| | - Claudia Angela Maria Fulgenzi
- Department of Surgery & Cancer, Imperial College London, UK.,Division of Medical Oncology, Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | | | | | | | - David J Pinato
- Department of Surgery & Cancer, Imperial College London, UK.,Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
23
|
Wheatley RC, Kilgour E, Jacobs T, Lamarca A, Hubner RA, Valle JW, McNamara MG. Potential influence of the microbiome environment in patients with biliary tract cancer and implications for therapy. Br J Cancer 2022; 126:693-705. [PMID: 34663949 PMCID: PMC8888758 DOI: 10.1038/s41416-021-01583-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/16/2021] [Accepted: 10/04/2021] [Indexed: 12/25/2022] Open
Abstract
Biliary tract cancers, including intra- and extra-hepatic cholangiocarcinoma as well as gallbladder cancer, are associated with poor prognosis and the majority of patients present with advanced-stage, non-resectable disease at diagnosis. Biliary tract cancer may develop through an accumulation of genetic and epigenetic alterations and can be influenced by microbial exposure. Furthermore, the liver and biliary tract are exposed to the gastrointestinal microbiome through the gut-liver axis. The availability of next-generation sequencing technology has led to an increase in studies investigating the relationship between microbiota and human disease. In particular, the interplay between the microbiome, the tumour micro-environment and response to systemic therapy is a prospering area of interest. Given the poor outcomes for patients with biliary tract cancer, this emerging field of research, through which new biomarkers may be identified, offers potential as a tool for early diagnosis, prognostication or even as a future therapeutic target. This review summarises the available evidence on the microbiome environment in patients with biliary tract cancer, including a discussion around confounding factors, implications for therapy and proposed future directions.
Collapse
Affiliation(s)
- Roseanna C Wheatley
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Elaine Kilgour
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Alderley Park, UK
| | - Timothy Jacobs
- The Library, The Christie NHS Foundation Trust, Manchester, UK
| | - Angela Lamarca
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Richard A Hubner
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Juan W Valle
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Mairéad G McNamara
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester, UK.
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK.
| |
Collapse
|
24
|
van de Velde ME, El Hassani SEM, Kaspers GJL, Broertjes J, Benninga MA, de Boer NKH, Budding AE, de Meij TGJ. Prediction of Bloodstream Infection in Pediatric Acute Leukemia by Microbiota and Volatile Organic Compounds Analysis. J Pediatr Hematol Oncol 2022; 44:e152-e159. [PMID: 34133377 DOI: 10.1097/mph.0000000000002210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 03/28/2021] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Bloodstream infections (BSIs) cause treatment-related mortality in pediatric acute leukemia. We explored the potential of intestinal microbiota and fecal volatile organic compounds (VOCs) analyses to predict BSI. METHODS In this case-control study, fecal samples of pediatric acute leukemia patients were collected. Microbiota composition and fecal VOC profiles of BSI cases and matched non-BSI controls were compared. RESULTS In total, 6 patients were included, of which 1 developed BSI and 1 neutropenic fever. Both showed reduced microbial diversity and stability of Bacteroidetes. In the BSI case, Pantoea was identified 15 days before BSI. Significant differences in fecal VOC profiles were measured between the case and controls. CONCLUSION Microbiota and fecal VOC could serve as biomarkers to predict BSI in pediatric leukemia.
Collapse
Affiliation(s)
| | - Sofia El Manouni El Hassani
- Pediatric Gastroenterology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam
- Department of Pediatric Gastroenterology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam
| | - Gert Jan L Kaspers
- Departments of Pediatric Oncology
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jorrit Broertjes
- Department of Pediatric Gastroenterology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam
| | - Marc A Benninga
- Department of Pediatric Gastroenterology, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam
| | - Nanne K H de Boer
- Department of Gastroenterology and Hepatology, Amsterdam UMC, Vrije Universiteit Amsterdam, AG&M Research Institute
| | | | - Tim G J de Meij
- Pediatric Gastroenterology, Emma Children's Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam
| |
Collapse
|
25
|
Balaian E, Wobus M, Bornhäuser M, Chavakis T, Sockel K. Myelodysplastic Syndromes and Metabolism. Int J Mol Sci 2021; 22:ijms222011250. [PMID: 34681910 PMCID: PMC8541058 DOI: 10.3390/ijms222011250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/06/2021] [Accepted: 10/14/2021] [Indexed: 12/01/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are acquired clonal stem cell disorders exhibiting ineffective hematopoiesis, dysplastic cell morphology in the bone marrow, and peripheral cytopenia at early stages; while advanced stages carry a high risk for transformation into acute myeloid leukemia (AML). Genetic alterations are integral to the pathogenesis of MDS. However, it remains unclear how these genetic changes in hematopoietic stem and progenitor cells (HSPCs) occur, and how they confer an expansion advantage to the clones carrying them. Recently, inflammatory processes and changes in cellular metabolism of HSPCs and the surrounding bone marrow microenvironment have been associated with an age-related dysfunction of HSPCs and the emergence of genetic aberrations related to clonal hematopoiesis of indeterminate potential (CHIP). The present review highlights the involvement of metabolic and inflammatory pathways in the regulation of HSPC and niche cell function in MDS in comparison to healthy state and discusses how such pathways may be amenable to therapeutic interventions.
Collapse
Affiliation(s)
- Ekaterina Balaian
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (M.W.); (M.B.)
- German Cancer Consortium (DKTK), Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Correspondence: (E.B.); (K.S.)
| | - Manja Wobus
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (M.W.); (M.B.)
| | - Martin Bornhäuser
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (M.W.); (M.B.)
- National Center for Tumor Diseases, Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Triantafyllos Chavakis
- National Center for Tumor Diseases, Partner Site Dresden and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Carl Gustav Carus Dresden, 01307 Dresden, Germany
| | - Katja Sockel
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany; (M.W.); (M.B.)
- Correspondence: (E.B.); (K.S.)
| |
Collapse
|
26
|
Mamgain G, Patra P, Naithani M, Nath UK. The Role of Microbiota in the Development of Cancer Tumour Cells and Lymphoma of B and T Cells. Cureus 2021; 13:e19047. [PMID: 34853760 PMCID: PMC8608681 DOI: 10.7759/cureus.19047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 11/26/2022] Open
Abstract
Human body harbours enormous numbers of microbial organisms, including bacteria, viruses, and fungi which have a momentous role in well-being and illness in humans. Immune system shelters us from pathogenic bacteria, microorganisms found in human tissues have many benefits related to the functional movement of the host by regulating important procedures such as immunity, signalling, and breakdown. Lymphocytes assume a significant part in the reaction to bacterial colonization, primarily by prompting a safe reaction to obstruction or initiation. Most immunologically occupant cells have a place with the mucosal invulnerable framework and are continually motioned by dendritic cells or other Antigen introducing cells that gather intestinal samples. Thus, Microbiome is a key contributor to developing lymphoma and specific alterations to microbiome composition could attenuate the risk. There is an indication that microbial morphology can affect and control humanoids. The difference in the composition of these microorganisms is associated with tumour development. With the increased knowledge of the connection among the human microbiome and carcinogenesis, the use of these findings to prevent, predict or diagnose of lymphomas has attracted a great attention. In this article, we explored current knowledge of various microbial ecosystems, their connection with carcinogens and the potential for useful microorganisms to control and prevent B and T cell lymphoma.
Collapse
Affiliation(s)
- Garima Mamgain
- Medical Oncology and Haematology, All India Institute of Medical Sciences, Rishikesh, IND
| | - Priyanka Patra
- Biochemistry, All India Institute of Medical Sciences, Rishikesh, IND
| | - Manisha Naithani
- Biochemistry & Advanced Center of Continuous Professional Development, All India Institute of Medical Sciences, Rishikesh, IND
| | - Uttam Kumar Nath
- Medical Oncology and Haematology, All India Institute of Medical Sciences, Rishikesh, IND
| |
Collapse
|
27
|
Oh B, Boyle F, Pavlakis N, Clarke S, Guminski A, Eade T, Lamoury G, Carroll S, Morgia M, Kneebone A, Hruby G, Stevens M, Liu W, Corless B, Molloy M, Libermann T, Rosenthal D, Back M. Emerging Evidence of the Gut Microbiome in Chemotherapy: A Clinical Review. Front Oncol 2021; 11:706331. [PMID: 34604043 PMCID: PMC8481611 DOI: 10.3389/fonc.2021.706331] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/25/2021] [Indexed: 01/28/2023] Open
Abstract
Increasing evidence suggests that the gut microbiome is associated with both cancer chemotherapy (CTX) outcomes and adverse events (AEs). This review examines the relationship between the gut microbiome and CTX as well as the impact of CTX on the gut microbiome. A literature search was conducted in electronic databases Medline, PubMed and ScienceDirect, with searches for "cancer" and "chemotherapy" and "microbiome/microbiota". The relevant literature was selected for use in this article. Seventeen studies were selected on participants with colorectal cancer (CRC; n=5), Acute Myeloid Leukemia (AML; n=3), Non-Hodgkin's lymphoma (n=2), breast cancer (BCa; n=1), lung cancer (n=1), ovarian cancer (n=1), liver cancer (n=1), and various other types of cancers (n=3). Seven studies assessed the relationship between the gut microbiome and CTX with faecal samples collected prior to (n=3) and following CTX (n=4) showing that the gut microbiome is associated with both CTX efficacy and toxicity. Ten other prospective studies assessed the impact of CTX during treatment and found that CTX modulates the gut microbiome of people with cancer and that dysbiosis induced by the CTX is related to AEs. CTX adversely impacts the gut microbiome, inducing dysbiosis and is associated with CTX outcomes and AEs. Current evidence provides insights into the gut microbiome for clinicians, cancer survivors and the general public. More research is required to better understand and modify the impact of CTX on the gut microbiome.
Collapse
Affiliation(s)
- Byeongsang Oh
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
- Cancer Care Service, Mater Hospital, North Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- University of Kansas Medical Center, Kansas City, KS, United States
| | - Frances Boyle
- Cancer Care Service, Mater Hospital, North Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Nick Pavlakis
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Stephen Clarke
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Alex Guminski
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
- Cancer Care Service, Mater Hospital, North Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Thomas Eade
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
- Cancer Care Service, Mater Hospital, North Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Gillian Lamoury
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
- Cancer Care Service, Mater Hospital, North Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Susan Carroll
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
- Cancer Care Service, Mater Hospital, North Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Marita Morgia
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
- Cancer Care Service, Mater Hospital, North Sydney, NSW, Australia
| | - Andrew Kneebone
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
- Cancer Care Service, Mater Hospital, North Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - George Hruby
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
- Cancer Care Service, Mater Hospital, North Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Mark Stevens
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
- Cancer Care Service, Mater Hospital, North Sydney, NSW, Australia
| | - Wen Liu
- University of Kansas Medical Center, Kansas City, KS, United States
| | - Brian Corless
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Mark Molloy
- Bowel Cancer and Biomarker Laboratory, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Towia Libermann
- Beth Israel Deaconess Medical Center (BIDMC) Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Harvard Medical School, Boston, MA, United States
| | | | - Michael Back
- Northern Sydney Cancer Centre, Royal North Shore Hospital, St Leonards, NSW, Australia
- Cancer Care Service, Mater Hospital, North Sydney, NSW, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
28
|
Yin T, Jeong JH, Hardcastle TF, Biswas K, Douglas RG. A scoping review of longitudinal airway microbiota studies. Expert Rev Respir Med 2021; 15:1187-1195. [PMID: 33908842 DOI: 10.1080/17476348.2021.1924061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 04/27/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION The role of the microbiota in inflammatory airway diseases is unclear. Antimicrobial therapies have predominantly been guided by culture results. However, molecular sequencing has shown that the airway microbiota is much more complex and accurate modeling requires longitudinal analysis. AREAS COVERED A Preferred Reporting Items for Systematic Reviews and Meta-Analyses scoping review was performed by searching Medline, Scopus, and Web of Science databases for all longitudinal airway microbiota studies that utilized molecular techniques. 38 studies with 1,993 participants were included in this review. Healthy microbial communities were more diverse, individualized and stable over time. Acute infections resulted in changes in the microbiota that were detected earlier and more sensitively by molecular sequencing than culture. Distinct microbiota profiles have been demonstrated in chronic obstructive pulmonary disease patients associated with exacerbation frequency and severity. EXPERT OPINION Longitudinal studies provide essential data on the stability of the microbiota over time and valuable information about the dynamic interactions between host, disease and microbes. We believe that molecular sequencing will be increasingly incorporated into research and clinical practice in the future. These advances can lead to improved diagnosis, enhanced prescribing guidance and reduce unnecessary antibiotic usage.
Collapse
Affiliation(s)
- Tary Yin
- Department of Surgery, The University of Auckland, Auckland, New Zealand
| | - Jae H Jeong
- Department of Otolaryngology-Head and Neck Surgery, Auckland District Health Board, Auckland, New Zealand
| | - Tim F Hardcastle
- Department of Otolaryngology-Head and Neck Surgery, Auckland District Health Board, Auckland, New Zealand
| | - Kristi Biswas
- Department of Surgery, The University of Auckland, Auckland, New Zealand
| | - Richard G Douglas
- Department of Surgery, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
29
|
D’Amico F, Perrone AM, Rampelli S, Coluccelli S, Barone M, Ravegnini G, Fabbrini M, Brigidi P, De Iaco P, Turroni S. Gut Microbiota Dynamics during Chemotherapy in Epithelial Ovarian Cancer Patients Are Related to Therapeutic Outcome. Cancers (Basel) 2021; 13:cancers13163999. [PMID: 34439153 PMCID: PMC8393652 DOI: 10.3390/cancers13163999] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/23/2021] [Accepted: 08/05/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary This pilot study on the trajectory of the gut microbiota (GM) in patients with epithelial ovarian cancer undergoing neoadjuvant and adjuvant chemotherapy highlighted peculiar dynamics associated with the therapeutic outcome. In particular, platinum-resistant patients showed a marked temporal reduction in GM diversity and increased instability with loss of health-associated taxa and increased proportions of lactate-producing microorganisms compared to those sensitive to platinum. These potential GM signatures of therapeutic failure are detectable within the first half of chemotherapy cycles, suggesting that early integrated treatments also aimed at modulating GM could influence therapeutic outcome. Further studies in larger cohorts combining multiple omics and possibly animal models are urgently needed for in-depth mechanistic understanding. Abstract Epithelial ovarian cancer (EOC) is one of the most lethal and silent gynecological tumors. Despite appropriate surgery and chemotherapy, relapse occurs in over half of patients with a poor prognosis. Recently, the gut microbiota (GM) was hypothesized to influence the efficacy of anticancer therapies, but no data are available in EOC. Here, by 16S rRNA gene sequencing and inferred metagenomics, we profiled the GM of EOC patients at diagnosis and reconstructed its trajectory along the course of neoadjuvant or adjuvant chemotherapy up to follow-up. Compared to healthy subjects, the GM of EOC patients appeared unbalanced and severely affected by chemotherapy. Strikingly, discriminating patterns were identified in relation to the therapeutic response. Platinum-resistant patients showed a marked temporal reduction in GM diversity and increased instability with loss of health-associated taxa and increased proportions of Coriobacteriaceae and Bifidobacterium. Notably, most of these microorganisms are lactate producers, suggesting increased lactate production as supported by inferred metagenomics. In contrast, the GM of platinum-sensitive patients appeared overall more diverse and stable and enriched in lactate utilizers from the Veillonellaceae family. In conclusion, we identified potential GM signatures of therapeutic outcome in EOC patients, which could open up new opportunities for cancer prognosis and treatment.
Collapse
Affiliation(s)
- Federica D’Amico
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (A.M.P.); (S.C.); (M.B.); (P.B.); (P.D.I.)
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (S.R.); (G.R.); (M.F.); (S.T.)
- Correspondence: ; Tel.: +39-051-2099727
| | - Anna Myriam Perrone
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (A.M.P.); (S.C.); (M.B.); (P.B.); (P.D.I.)
- Division of Oncologic Gynecology, IRCCS Azienda Ospedaliero, University of Bologna, 40138 Bologna, Italy
- Centro di Studio e Ricerca delle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
| | - Simone Rampelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (S.R.); (G.R.); (M.F.); (S.T.)
| | - Sara Coluccelli
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (A.M.P.); (S.C.); (M.B.); (P.B.); (P.D.I.)
- Division of Oncologic Gynecology, IRCCS Azienda Ospedaliero, University of Bologna, 40138 Bologna, Italy
| | - Monica Barone
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (A.M.P.); (S.C.); (M.B.); (P.B.); (P.D.I.)
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (S.R.); (G.R.); (M.F.); (S.T.)
| | - Gloria Ravegnini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (S.R.); (G.R.); (M.F.); (S.T.)
| | - Marco Fabbrini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (S.R.); (G.R.); (M.F.); (S.T.)
| | - Patrizia Brigidi
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (A.M.P.); (S.C.); (M.B.); (P.B.); (P.D.I.)
- Centro di Studio e Ricerca delle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
| | - Pierandrea De Iaco
- Department of Medical and Surgical Sciences, University of Bologna, 40138 Bologna, Italy; (A.M.P.); (S.C.); (M.B.); (P.B.); (P.D.I.)
- Division of Oncologic Gynecology, IRCCS Azienda Ospedaliero, University of Bologna, 40138 Bologna, Italy
- Centro di Studio e Ricerca delle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (S.R.); (G.R.); (M.F.); (S.T.)
- Centro di Studio e Ricerca delle Neoplasie Ginecologiche (CSR), University of Bologna, 40138 Bologna, Italy
| |
Collapse
|
30
|
An infectious diseases perspective on the microbiome and allogeneic stem cell transplant. Curr Opin Infect Dis 2021; 33:426-432. [PMID: 33148984 DOI: 10.1097/qco.0000000000000683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
PURPOSE OF REVIEW The gut microbiome presents a novel source of diagnostic and therapeutic potential to modify post allogeneic stem cell transplant complications. There is an explosion of interest in microbiome research, mostly in the form of single-centre prospective time-series cohorts utilizing a variety of sampling frequencies and metagenomic technologies to sequence the microbiome. The purpose of this review is to summarize important recent publications and contextualize them within what has already been described in this rapidly growing field. RECENT FINDING Results from observational human cohort and animal transplant models add to the growing body of evidence that the microbiome modulates the immunopathogenesis of posttransplant complications. This is particularly the case for recipients of grafts replete with T cells where the evidence that acute graft-versus-host disease is mediated by anaerobic commensal-associated short-chain fatty acids, which interact with mucosa-associated (CD4FOXP3) T-regulatory cells. SUMMARY Future human research into the role of the microbiome in allogeneic stem transplant should incorporate rigorous and considered experimental design in addition to next-generation sequencing technology to better portray microbiome functional potential and active gene expression. In combination with host immune phenotyping, which would facilitate a robust understanding of the host--microbiome interaction that is required before meaningful translation into clinical diagnostics and therapeutics can be expected.
Collapse
|
31
|
Shuler K, Verbanic S, Chen IA, Lee J. A Bayesian nonparametric analysis for zero‐inflated multivariate count data with application to microbiome study. J R Stat Soc Ser C Appl Stat 2021. [DOI: 10.1111/rssc.12493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Kurtis Shuler
- Sandia National Laboratories in Albuquerque Albuquerque NM USA
| | - Samuel Verbanic
- Department of Chemical and Biomolecular Engineering University of California Los Angeles Los Angeles CA USA
| | - Irene A. Chen
- Department of Chemical and Biomolecular Engineering University of California Los Angeles Los Angeles CA USA
| | - Juhee Lee
- Department of Statistics University of California Santa Cruz Santa Cruz CA USA
| |
Collapse
|
32
|
Malard F, Vekhoff A, Lapusan S, Isnard F, D'incan-Corda E, Rey J, Saillard C, Thomas X, Ducastelle-Lepretre S, Paubelle E, Larcher MV, Rocher C, Recher C, Tavitian S, Bertoli S, Michallet AS, Gilis L, Peterlin P, Chevallier P, Nguyen S, Plantamura E, Boucinha L, Gasc C, Michallet M, Dore J, Legrand O, Mohty M. Gut microbiota diversity after autologous fecal microbiota transfer in acute myeloid leukemia patients. Nat Commun 2021; 12:3084. [PMID: 34035290 PMCID: PMC8149453 DOI: 10.1038/s41467-021-23376-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 04/22/2021] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) intensive chemotherapy combined with broad-spectrum antibiotics, leads to gut microbiota dysbiosis promoting pathological conditions and an increased incidence of complications. Here we report findings from a phase II single-arm, multicenter study evaluating autologous fecal microbiota transfer (AFMT) in 25 AML patients treated with intensive chemotherapy and antibiotics (ClinicalTrials.gov number: NCT02928523). The co-primary outcomes of the study are to evaluate the efficacy of AFMT in dysbiosis correction and multidrug-resistant bacteria eradication. The main secondary outcomes are to define a dysbiosis biosignature, to evaluate the effect of dysbiosis correction on patient clinical status, to assess the short and mid-term safety of AFMT in this immunocompromised population, and to evaluate the feasibility of the AFMT procedure and acceptability by the patient. Intensive induction chemotherapy induces a dramatic decrease of α-diversity indices, and a microbial dysbiosis with a significant shift of the microbial communities and domination of pro-inflammatory families. After AFMT treatment, α-diversity indices return to their initial mean levels and the similarity index shows the restoration of microbial communities. The trial meets pre-specified endpoints. AFMT appears to be safe and may be effective for gut microbiota restoration in AML patients receiving intensive chemotherapy and antibiotics, with an excellent gut microbiota reconstruction based on both richness and diversity indices at the species level. The combination of chemotherapy and broad-spectrum antibiotics induces gut microbiota (GM) dysbiosis in acute myeloid leukaemia (AML) leading to additional complications. Here, the authors report the efficacy in GM restoration and safety of autologous faecal microbiota transfer in treated AML patients in a phase II clinical trial.
Collapse
Affiliation(s)
- Florent Malard
- Service d'hématologie clinique et de thérapie cellulaire, Hôpital Saint Antoine, APHP, Sorbonne Université, INSERM UMRs 938, Paris, France.
| | - Anne Vekhoff
- Service d'hématologie clinique et de thérapie cellulaire, Hôpital Saint Antoine, APHP, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - Simona Lapusan
- Service d'hématologie clinique et de thérapie cellulaire, Hôpital Saint Antoine, APHP, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - Francoise Isnard
- Service d'hématologie clinique et de thérapie cellulaire, Hôpital Saint Antoine, APHP, Sorbonne Université, INSERM UMRs 938, Paris, France
| | | | - Jérôme Rey
- Service d'hématologie, Institut Paoli Calmettes, Marseille, France
| | - Colombe Saillard
- Service d'hématologie, Institut Paoli Calmettes, Marseille, France
| | - Xavier Thomas
- Service d'hématologie, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | | | - Etienne Paubelle
- Service d'hématologie, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Marie-Virginie Larcher
- Service d'hématologie, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Clément Rocher
- Service d'hématologie, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France
| | - Christian Recher
- CHU de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Université de Toulouse III Paul Sabatier, Service d'hématologie, Toulouse, France
| | - Suzanne Tavitian
- CHU de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Université de Toulouse III Paul Sabatier, Service d'hématologie, Toulouse, France
| | - Sarah Bertoli
- CHU de Toulouse, Institut Universitaire du Cancer de Toulouse Oncopole, Université de Toulouse III Paul Sabatier, Service d'hématologie, Toulouse, France
| | | | - Lila Gilis
- Service d'hématologie, Centre Léon Bérard, Lyon, France
| | | | | | - Stéphanie Nguyen
- Service d'hématologie clinique, Hôpital de la Pitié Salpétrière, APHP, Sorbonne Université, Paris, France
| | | | | | | | | | - Joel Dore
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS, Jouy-en-Josas, France
| | - Ollivier Legrand
- Service d'hématologie clinique et de thérapie cellulaire, Hôpital Saint Antoine, APHP, Sorbonne Université, INSERM UMRs 938, Paris, France
| | - Mohamad Mohty
- Service d'hématologie clinique et de thérapie cellulaire, Hôpital Saint Antoine, APHP, Sorbonne Université, INSERM UMRs 938, Paris, France
| |
Collapse
|
33
|
Khan N, Lindner S, Gomes ALC, Devlin SM, Shah GL, Sung AD, Sauter CS, Landau HJ, Dahi PB, Perales MA, Chung DJ, Lesokhin AM, Dai A, Clurman A, Slingerland JB, Slingerland AE, Brereton DG, Giardina PA, Maloy M, Armijo GK, Rondon-Clavo C, Fontana E, Bohannon L, Ramalingam S, Bush AT, Lew MV, Messina JA, Littmann E, Taur Y, Jenq RR, Chao NJ, Giralt S, Markey KA, Pamer EG, van den Brink MRM, Peled JU. Fecal microbiota diversity disruption and clinical outcomes after auto-HCT: a multicenter observational study. Blood 2021; 137:1527-1537. [PMID: 33512409 PMCID: PMC7976512 DOI: 10.1182/blood.2020006923] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 11/09/2020] [Indexed: 12/18/2022] Open
Abstract
We previously described clinically relevant reductions in fecal microbiota diversity in patients undergoing allogeneic hematopoietic cell transplantation (allo-HCT). Recipients of high-dose chemotherapy and autologous HCT (auto-HCT) incur similar antibiotic exposures and nutritional alterations. To characterize the fecal microbiota in the auto-HCT population, we analyzed 1161 fecal samples collected from 534 adult recipients of auto-HCT for lymphoma, myeloma, and amyloidosis in an observational study conducted at 2 transplantation centers in the United States. By using 16S ribosomal gene sequencing, we assessed fecal microbiota composition and diversity, as measured by the inverse Simpson index. At both centers, the diversity of early pretransplant fecal microbiota was lower in patients than in healthy controls and decreased further during the course of transplantation. Loss of diversity and domination by specific bacterial taxa occurred during auto-HCT in patterns similar to those with allo-HCT. Above-median fecal intestinal diversity in the periengraftment period was associated with decreased risk of death or progression (progression-free survival hazard ratio, 0.46; 95% confidence interval, 0.26-0.82; P = .008), adjusting for disease and disease status. This suggests that further investigation into the health of the intestinal microbiota in auto-HCT patients and posttransplant outcomes should be undertaken.
Collapse
Affiliation(s)
- Niloufer Khan
- Adult Bone Marrow Transplantation Service, Department of Medicine
| | - Sarah Lindner
- Department of Immunology, Sloan Kettering Institute, and
| | | | - Sean M Devlin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Gunjan L Shah
- Adult Bone Marrow Transplantation Service, Department of Medicine
- Weill Cornell Medical College, New York, NY
| | - Anthony D Sung
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Craig S Sauter
- Adult Bone Marrow Transplantation Service, Department of Medicine
- Weill Cornell Medical College, New York, NY
| | - Heather J Landau
- Adult Bone Marrow Transplantation Service, Department of Medicine
- Weill Cornell Medical College, New York, NY
| | - Parastoo B Dahi
- Adult Bone Marrow Transplantation Service, Department of Medicine
- Weill Cornell Medical College, New York, NY
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine
- Weill Cornell Medical College, New York, NY
| | - David J Chung
- Adult Bone Marrow Transplantation Service, Department of Medicine
- Weill Cornell Medical College, New York, NY
| | - Alexander M Lesokhin
- Weill Cornell Medical College, New York, NY
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anqi Dai
- Department of Immunology, Sloan Kettering Institute, and
| | - Annelie Clurman
- Adult Bone Marrow Transplantation Service, Department of Medicine
| | | | | | | | - Paul A Giardina
- Adult Bone Marrow Transplantation Service, Department of Medicine
| | - Molly Maloy
- Adult Bone Marrow Transplantation Service, Department of Medicine
| | | | | | - Emily Fontana
- Department of Immunology, Sloan Kettering Institute, and
| | - Lauren Bohannon
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Sendhilnathan Ramalingam
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Amy T Bush
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Meagan V Lew
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Julia A Messina
- Division of Infectious Diseases, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Eric Littmann
- Duchossois Family Institute, University of Chicago, Chicago, IL
| | - Ying Taur
- Weill Cornell Medical College, New York, NY
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY; and
| | - Robert R Jenq
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nelson J Chao
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, Duke University School of Medicine, Durham, NC
| | - Sergio Giralt
- Adult Bone Marrow Transplantation Service, Department of Medicine
- Weill Cornell Medical College, New York, NY
| | - Kate A Markey
- Adult Bone Marrow Transplantation Service, Department of Medicine
- Weill Cornell Medical College, New York, NY
| | - Eric G Pamer
- Duchossois Family Institute, University of Chicago, Chicago, IL
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine
- Department of Immunology, Sloan Kettering Institute, and
- Weill Cornell Medical College, New York, NY
| | - Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine
- Weill Cornell Medical College, New York, NY
| |
Collapse
|
34
|
Characterization of microbiota in acute leukemia patients following successful remission induction chemotherapy without antimicrobial prophylaxis. Int Microbiol 2021; 24:263-273. [DOI: 10.1007/s10123-021-00163-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/10/2021] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
|
35
|
Gut Microbiota Influence in Hematological Malignancies: From Genesis to Cure. Int J Mol Sci 2021; 22:ijms22031026. [PMID: 33498529 PMCID: PMC7864170 DOI: 10.3390/ijms22031026] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/13/2022] Open
Abstract
Hematological malignancies, including multiple myeloma, lymphoma, and leukemia, are a heterogeneous group of neoplasms that affect the blood, bone marrow, and lymph nodes. They originate from uncontrolled growth of hematopoietic and lymphoid cells from different stages in their maturation/differentiation and account for 6.5% of all cancers around the world. During the last decade, it has been proven that the gut microbiota, more specifically the gastrointestinal commensal bacteria, is implicated in the genesis and progression of many diseases. The immune-modulating effects of the human microbiota extend well beyond the gut, mostly through the small molecules they produce. This review aims to summarize the current knowledge of the role of the microbiota in modulating the immune system, its role in hematological malignancies, and its influence on different therapies for these diseases, including autologous and allogeneic stem cell transplantation, chemotherapy, and chimeric antigen receptor T cells.
Collapse
|
36
|
Oral Health, Caries Risk Profiles, and Oral Microbiome of Pediatric Patients with Leukemia Submitted to Chemotherapy. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6637503. [PMID: 33532491 PMCID: PMC7834790 DOI: 10.1155/2021/6637503] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/29/2020] [Accepted: 01/06/2021] [Indexed: 02/05/2023]
Abstract
Background Chemotherapy is the primary treatment modality used for patients with acute lymphoblastic leukemia (ALL), but inevitably causes microbiota-related oral complications. This study is aimed at investigating the effects of chemotherapy on oral health status, caries risk, and oral microbiome in pediatric patients with ALL. Methods Thirty-nine children with ALL receiving chemotherapy were enrolled, and a gender-, age-, dentition stage, and socioeconomic class matched healthy counterpart were recruited. Demographic information and overall health condition were obtained through the questionnaire and medical records. Oral examination was performed to assess caries and salivary status, plaque index, and other oral manifestations. Cariogram was used to assess the overall caries risk. Supragingival samples of thirteen ALL subjects and their counterparts were randomly selected to perform a 16S ribosomal RNA gene 454 pyrosequencing. Raw sequence data were screened, trimmed, and filtered using Seqcln and MOTHUR. Results The prevalence of dental caries, gingivitis, oral mucositis, xerostomia, and candidiasis in ALL groups was higher than that of the control group (p < 0.05). Children with ALL demonstrated higher caries risk compared to healthy controls (HC) based upon Cariogram (p < 0.05). The oral microbial structure of ALL patients receiving chemotherapy is different from that of healthy controls. Oral microbiota of ALL groups showed less alpha diversity and significant differences in the composition of the oral microbiome compared to healthy controls. Conclusions ALL patients receiving chemotherapy demonstrated compromised oral health, high caries risk, alteration of caries-related factors, and dysbiosis of oral microbiota. These findings may be of clinical importance in developing better strategies for personalized preventive management of oral diseases for pediatric children with ALL.
Collapse
|
37
|
Mougeot JLC, Beckman MF, Langdon HC, Brennan MT, Bahrani Mougeot F. Oral Microbiome Signatures in Hematological Cancers Reveal Predominance of Actinomyces and Rothia Species. J Clin Med 2020; 9:jcm9124068. [PMID: 33348567 PMCID: PMC7767039 DOI: 10.3390/jcm9124068] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
The endogenous microbiome of healthy individuals in oral cavities is diverse, representing over 700 bacterial species. Imbalance in oral and gut microbiome composition and associated gene expression has been linked to different forms of hematological (blood) cancers. Our objective is to compare oral microbiome profiles of patients with blood cancers (BC group: N = 39 patients, n = 124 oral samples) to those of healthy control subjects (HC group: N = 27 subjects, n = 100 oral samples). Saliva samples and swabs of buccal mucosa, supragingival plaque, and tongue were collected from blood cancer patients and healthy controls. Next-generation sequencing (16S-rRNA gene V3-V4 region) was used to determine the relative abundance of bacterial taxa present at the genus and species levels. Differences in oral microbiome beta-diversity were determined using multivariate permutational analysis of variance (PERMANOVA). Linear discriminant analysis (LDA) effect size (LEfSe) analysis was performed to identify differentiating bacterial taxa in pairwise comparisons. The PATRICv3.6.7 online tool was used to determine the predominance of potential pathogenicity in the BC group. The oral microbiome beta-diversities of the BC and HC groups differed and corresponded to a reduced alpha-diversity in the BC group. LEfSe analysis showed significant LDA scores for Actinomyces and Rothia spp., differentiating the BC group from the HC group. In silico analysis using PATRICv3.6.7 demonstrated that the groups of bacteria possessing traits of "antibiotic resistance", "oral pathogen", and "virulence" was enriched in the BC group. Although 56% of the BC patients received antibiotics within two weeks of the oral bacterial sampling, Actinomyces genus remained the top differentiating feature in the BC group regardless of the administration of antibiotics, while Rothia dentocariosa was detected as the top differentiating feature in the BC patients who did not receive antibiotics, but not in those who received antibiotics. Further investigation is needed to better understand the interactions of certain oral species with the host immune system to better characterize clinically relevant associations with hematological cancers.
Collapse
Affiliation(s)
- Jean-Luc C. Mougeot
- Correspondence: (J.-L.C.M.); (F.B.M.); Tel.: +1-704-355-5301 (J.-L.C.M.); +1-704-355-8132 (F.B.M.)
| | | | | | | | - Farah Bahrani Mougeot
- Correspondence: (J.-L.C.M.); (F.B.M.); Tel.: +1-704-355-5301 (J.-L.C.M.); +1-704-355-8132 (F.B.M.)
| |
Collapse
|
38
|
Zhao Y, Wang C, Goel A. Role of gut microbiota in epigenetic regulation of colorectal Cancer. Biochim Biophys Acta Rev Cancer 2020; 1875:188490. [PMID: 33321173 DOI: 10.1016/j.bbcan.2020.188490] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/02/2020] [Accepted: 12/02/2020] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) remains one of the most commonly diagnosed cancers and a leading cause of cancer-related deaths worldwide. The stepwise accumulation of epigenetic alterations in the normal colorectal epithelium has been reported to act as a driving force for the initiation and promotion of tumorigenesis in CRC. From a mechanistic standpoint, emerging evidence indicates that within the colorectal epithelium, the diverse gut microbiota can interact with host cells to regulate multiple physiological processes. In fact, recent studies have found that the gut microbiota represents a potential cause of carcinogenesis, invasion, and metastasis via DNA methylation, histone modifications, and non-coding RNAs - providing an epigenetic perspective for the connection between the gut microbiota and CRC. Herein, we comprehensively review the recent research that provides a comprehensive yet succinct evidence connecting the gut microbiota to CRC at an epigenetic level, including carcinogenic mechanisms of cancer-related microbiota, and the potential for utilizing the gut microbiota as CRC biomarkers. These scientific findings highlight a promising future for manipulating the gut microbiota to improve clinical outcomes in patients suffering from CRC.
Collapse
Affiliation(s)
- Yinghui Zhao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China; Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China; Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China; Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, USA.
| |
Collapse
|
39
|
Innao V, Allegra AG, Musolino C, Allegra A. New Frontiers about the Role of Human Microbiota in Immunotherapy: The Immune Checkpoint Inhibitors and CAR T-Cell Therapy Era. Int J Mol Sci 2020; 21:ijms21238902. [PMID: 33255336 PMCID: PMC7727716 DOI: 10.3390/ijms21238902] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Microbiota is considered an independent organ with the capability to modulate tumor growth and response to therapies. In the chemo-free era, the use of new immunotherapies, more selective and effective and less toxic, led to the extension of overall survival of patients, subject to their ability to not stop treatment. This has focused scientists’ attention to optimize responses by understanding and changing microbiota composition. While we have obtained abundant data from studies in oncologic and hematologic patients receiving conventional chemotherapy, we have less data about alterations in intestinal flora in those undergoing immunotherapy, especially based on Chimeric Antigen Receptor (CAR) T-cells. Actually, we know that the efficacy of Programmed Cell Death 1 (PD-1), PD-1 ligand, and Cytotoxic T lymphocyte-associated protein 4 (CTLA-4) is improved by probiotics rich in Bifidobacterium spp., while compounds of Bacteroidales and Burkholderiales protect from the development of the anti-CTLA-4-induced colitis in mouse models. CAR T-cell therapy seems to not be interfering with microbiota; however, the numerous previous therapies may have caused permanent damage, thus obscuring the data we might have obtained. Therefore, this review opens a new chapter to transfer known acquisitions to a typology of patients destined to grow.
Collapse
Affiliation(s)
- Vanessa Innao
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood, University of Messina, 98122 Messina, Italy;
- Correspondence: (V.I.); (A.A.)
| | - Andrea Gaetano Allegra
- Radiation Oncology Unit, Department of Biomedical, Experimental, and Clinical Sciences “Mario Serio”, Azienda Ospedaliero-Universitaria Careggi, University of Florence, 50100 Florence, Italy;
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood, University of Messina, 98122 Messina, Italy;
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood, University of Messina, 98122 Messina, Italy;
- Correspondence: (V.I.); (A.A.)
| |
Collapse
|
40
|
Rund D. Intravenous iron: do we adequately understand the short- and long-term risks in clinical practice? Br J Haematol 2020; 193:466-480. [PMID: 33216989 DOI: 10.1111/bjh.17202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 09/28/2020] [Indexed: 12/31/2022]
Abstract
Intravenous (IV) iron as a therapeutic agent is often administered but not always fully understood. The benefits of IV iron are well proven in many fields, particularly in nephrology. IV iron is beneficial not only for true iron deficiency but also for iron-restricted anaemia (functional iron deficiency). Yet, the literature on intravenous iron has many inconsistencies regarding its adverse effects. Over the last several years, newer forms of iron have been developed, leading to the more regular use of iron and in larger doses. This review will summarize some of the older and newer literature regarding the differences among iron products, including the mechanisms and frequency of their adverse events (AEs). The pathway and frequency of an underrecognized adverse event (hypophosphataemia) will be discussed. Recent insights on infection risk and iron handling by macrophages are examined. Potential but presently unproven risks of iron overload due to IV iron are discussed. The impact of these on the risk:benefit ratio and dosing of intravenous iron are considered in different clinical settings, including pregnancy and cancer. IV iron is an essential component of the therapy of anaemia and understanding these issues will enable more informed treatment decisions and knowledgeable use of these drugs.
Collapse
Affiliation(s)
- Deborah Rund
- Hebrew University-Hadassah Medical Organization, Ein Kerem, Jerusalem, Israel
| |
Collapse
|
41
|
Ma C, Wasti S, Huang S, Zhang Z, Mishra R, Jiang S, You Z, Wu Y, Chang H, Wang Y, Huo D, Li C, Sun Z, Sun Z, Zhang J. The gut microbiome stability is altered by probiotic ingestion and improved by the continuous supplementation of galactooligosaccharide. Gut Microbes 2020; 12:1785252. [PMID: 32663059 PMCID: PMC7524268 DOI: 10.1080/19490976.2020.1785252] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The stable gut microbiome plays a key role in sustaining host health, while the instability of gut microbiome also has been found to be a risk factor of various metabolic diseases. At the ecological and evolutionary scales, the inevitable competition between the ingested probiotic and indigenous gut microbiome can lead to an increase in the instability. It remains largely unclear if and how exogenous prebiotic can improve the overall gut microbiome stability in probiotic consumption. In this study, we used Lactobacillus plantarum HNU082 (Lp082) as a model probiotic to examine the impact of the continuous or pulsed supplementation of galactooligosaccharide (GOS) on the gut microbiome stability in mice using shotgun metagenomic sequencing. Only continuous GOS supplement promoted the growth of probiotic and decreased its single-nucleotide polymorphisms (SNPs) mutation under competitive conditions. Besides, persistent GOS supplementation increased the overall stability, reshaped the probiotic competitive interactions with Bacteroides species in the indigenous microbiome, which was also evident by over-abundance of carbohydrate-active enzymes (CAZymes) accordingly. Also, we identified a total of 793 SNPs arisen in probiotic administration in the indigenous microbiome. Over 90% of them derived from Bacteroides species, which involved genes encoding transposase, CAZymes, and membrane proteins. However, neither GOS supplementation here de-escalated the overall adaptive mutations within the indigenous microbes during probiotic intake. Collectively, our study demonstrated the beneficial effect of continuous prebiotic supplementation on the ecological and genetic stability of gut microbiomes.
Collapse
Affiliation(s)
- Chenchen Ma
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Sanjeev Wasti
- Department of Human Nutrition, Food and Animal Science, University of Hawaii, Honolulu, HI, USA
| | - Shi Huang
- Single-Cell Center, CAS Key Laboratory of Biofuels and Shandong Key Laboratory of Energy Genetics, Qingdao Institute of BioEnergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China
| | - Zeng Zhang
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Rajeev Mishra
- Department of Human Nutrition, Food and Animal Science, University of Hawaii, Honolulu, HI, USA
| | - Shuaiming Jiang
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Zhengkai You
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Yixuan Wu
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Haibo Chang
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Yuanyuan Wang
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Dongxue Huo
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Congfa Li
- College of Food Science and Engineering, Hainan University, Haikou, China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education P. R. C., Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs China, Inner Mongolia Agricultural University, Hohhot, China,Zhihong Sun Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education P. R. C., Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs China, Inner Mongolia Agricultural University, Hohhot, Inner mongolia Autonomous Region, 010018, China
| | - Zheng Sun
- Single-Cell Center, CAS Key Laboratory of Biofuels and Shandong Key Laboratory of Energy Genetics, Qingdao Institute of BioEnergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, China,Zheng Sun Single-Cell Center, CAS Key Laboratory of Biofuels and Shandong Key Laboratory of Energy Genetics, Qingdao Institute of BioEnergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao, Shandong, 266101, China
| | - Jiachao Zhang
- College of Food Science and Engineering, Hainan University, Haikou, China,CONTACT Jiachao Zhang College of Food Science and Engineering, Hainan University, Haikou, Hainan570228, China
| |
Collapse
|
42
|
Allogenic stem cell transplant-associated acute graft versus host disease: a computational drug discovery text mining approach using oral and gut microbiome signatures. Support Care Cancer 2020; 29:1765-1779. [PMID: 33094358 DOI: 10.1007/s00520-020-05821-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/07/2020] [Indexed: 12/23/2022]
Abstract
PURPOSE Acute graft versus host disease (aGVHD) is a major cause of non-relapse morbidity and mortality post-allogenic hematopoietic stem cell transplant (HSCT). Using conventional literature search and computational approaches, our objective was to identify oral and gut bacterial species associated with aGVHD, potentially affecting drug treatment via lipopolysaccharide (LPS) pathways. METHODS Medline, PubMed, PubMed Central, and Google Scholar were searched using MeSH terms. The top 100 hits per database were curated, and 25 research articles were selected to examine oral and gut microbiomes associated with health, HSCT, and aGVHD. Literature search validation, aGVHD drug targets, and microbial metabolic pathway identification were completed using BioReader, MACADAM, KEGG, and STRING programs. RESULTS Our review determined that (1) oral genera Rothia, Solobacterium, and Veillonella were identified in HSCT patients' stool and associated with aGVHD; (2) shifts in gut enterococci profiles were determined in HSCT-associated aGVHD; (3) gut microbiome dysbiosis prior or during HSCT and lower Shannon diversity index at time of HSCT were also associated with increased risk of aGVHD and transplant related death; and (4) Coriobacteriaceae family was negatively correlated with gut aGVHD, whereas Eubacterium limosum was associated with decreased risk of chronic GVHD relapse. Additionally, we identified molecular pathways related to TLR4/ LPS, including candidate aGVHD drug targets, impacted by oral and gut bacterial taxa. CONCLUSION Reduced microbial diversity reflects higher severity and mortality rate in HSCT patients with aGVHD. Multi-omics approaches to decipher oral and gut microbiome associations will be critical for developing aGVHD preventive therapies.
Collapse
|
43
|
Belknap KC, Cote AL, McGill CM, Andam CP, Barth BM. The Role of the Microbiome in Cancer and the Development of Cancer Therapeutics. INTERNATIONAL JOURNAL OF BIOPHARMACEUTICAL SCIENCES 2020; 2:118. [PMID: 33778816 PMCID: PMC7993822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cancer is caused by a compilation of hereditary and environmental factors. In the past decade, next-generation sequencing has revealed the extent to which the microbiome influences the maintenance of homeostasis and therefore the prevention of diseases such as cancer. Current research efforts explore the interaction between cancer and the microbiome, and the results are anticipated to transform how clinicians approach cancer treatment. There is a plausible transition from the use of human genetic biomarkers to microbiomic biomarkers for genomic diagnostics. Considering the expanding knowledge of the ways in which the microbiome can affect the development of cancer, clinicians treating cancer patients should be considerate of how the microbiome can influence the host-drug or microbiome-cancer interactions. Recognition of the importance of the microbiome within the field of oncology is pertinent to understanding and furthering cancer development and treatment.
Collapse
Affiliation(s)
- Kaitlyn C. Belknap
- Department of Molecular, Cellular and Biomedical Sciences,
University of New Hampshire, Durham NH 03824 USA
| | - Andrea L. Cote
- Department of Molecular, Cellular and Biomedical Sciences,
University of New Hampshire, Durham NH 03824 USA
| | - Colin M. McGill
- Department of Chemistry, University of Alaska Anchorage,
Anchorage AK 99508 USA
| | - Cheryl P. Andam
- Department of Molecular, Cellular and Biomedical Sciences,
University of New Hampshire, Durham NH 03824 USA
- Department of Biological Sciences, University at Albany,
State University of New York, Albany, NY 12222 USA
| | - Brian M. Barth
- Department of Molecular, Cellular and Biomedical Sciences,
University of New Hampshire, Durham NH 03824 USA
| |
Collapse
|
44
|
Galloway-Peña JR, Shi Y, Peterson CB, Sahasrabhojane P, Gopalakrishnan V, Brumlow CE, Daver NG, Alfayez M, Boddu PC, Khan MAW, Wargo JA, Do KA, Jenq RR, Kontoyiannis DP, Shelburne SA. Gut Microbiome Signatures Are Predictive of Infectious Risk Following Induction Therapy for Acute Myeloid Leukemia. Clin Infect Dis 2020; 71:63-71. [PMID: 31436833 PMCID: PMC7312220 DOI: 10.1093/cid/ciz777] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 08/20/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The majority of studies that provide insights into the influence of the microbiome on the health of hematologic malignancy patients have concentrated on the transplant setting. Here, we sought to assess the predictive capacity of the gastrointestinal microbiome and its relationship to infectious outcomes in patients with acute myeloid leukemia (AML). METHODS 16s rRNA-based analysis was performed on oral swabs and stool samples obtained biweekly from baseline until neutrophil recovery following induction chemotherapy (IC) in 97 AML patients. Microbiome characteristics were correlated with clinical outcomes both during and after IC completion. RESULTS At the start of IC, higher stool Shannon diversity (hazard ratio [HR], 0.36; 95% confidence interval [CI], .18-.74) and higher relative abundance of Porphyromonadaceae (HR, 0.36; 95% CI, .18-.73) were associated with increased probability of remaining infection-free during neutropenia. A baseline stool Shannon diversity cutoff of <2 had optimal operating characteristics for predicting infectious complications during neutropenia. Although 56 patients received therapy >72 hours with a carbapenem, none of the patients had an infection with an extended spectrum β-lactamase-producing organism. Patients who received carbapenems for >72 hours had significantly lower α-diversity at neutrophil recovery (P = .001) and were approximately 4 times more likely to have infection in the 90 days following neutrophil recovery (HR, 4.55; 95% CI, 1.73-11.93). CONCLUSIONS Our results suggest that gut microbiome evaluation could assist with infectious risk stratification and that improved targeting of antibiotic administration during IC could decrease subsequent infectious complications in AML patients.Baseline microbiome diversity is a strong independent predictor of infection during acute myeloid leukemia induction chemotherapy (IC) among clinical and microbiome covariates. Higher baseline levels of Porphyromonadaceae appear protective against infection, while carbapenem use is associated with consequences to the microbiome and infection susceptibility post-IC.
Collapse
Affiliation(s)
- Jessica R Galloway-Peña
- Department of Genomic Medicine, Houston, Texas, USA
- Department of Infectious Disease, Infection Control, and Employee Health, Houston, Texas, USA
| | - Yushu Shi
- Department of Biostatistics, Houston, Texas, USA
| | | | - Pranoti Sahasrabhojane
- Department of Infectious Disease, Infection Control, and Employee Health, Houston, Texas, USA
| | | | - Chelcy E Brumlow
- Department of Infectious Disease, Infection Control, and Employee Health, Houston, Texas, USA
| | - Naval G Daver
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Mansour Alfayez
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Prajwal C Boddu
- Department of Hematology-Oncology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | - Jennifer A Wargo
- Department of Genomic Medicine, Houston, Texas, USA
- Department of Surgical Oncology, Houston, Texas, USA
| | - Kim-Anh Do
- Department of Biostatistics, Houston, Texas, USA
| | - Robert R Jenq
- Department of Genomic Medicine, Houston, Texas, USA
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Samuel A Shelburne
- Department of Genomic Medicine, Houston, Texas, USA
- Department of Infectious Disease, Infection Control, and Employee Health, Houston, Texas, USA
| |
Collapse
|
45
|
Rashidi A, Weisdorf DJ. Microbiota-based approaches to mitigate infectious complications of intensive chemotherapy in patients with acute leukemia. Transl Res 2020; 220:167-181. [PMID: 32275896 PMCID: PMC7605891 DOI: 10.1016/j.trsl.2020.03.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/03/2020] [Accepted: 03/24/2020] [Indexed: 12/12/2022]
Abstract
Despite advances in antimicrobial treatments, infection remains a common complication of intensive chemotherapy in patients with acute leukemia. It has become progressively apparent that the current antimicrobial focus has shortcomings that result from disruption of the commensal microbial communities of the gut. These effects, collectively known as dysbiosis, have been increasingly associated worldwide with growing complications such as Clostridioides difficile infection, systemic infections, and antibiotic resistance. A revision of the current practice is overdue. Several innovative concepts have been proposed and tested in animal models and humans, with the overarching goal of preventing damage to the microbiota and facilitating its recovery. In this review, we discuss these approaches, examine critical knowledge gaps, and explore how they may be filled in future research.
Collapse
Affiliation(s)
- Armin Rashidi
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.
| | - Daniel J Weisdorf
- Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
46
|
Observational Cohort Study of Oral Mycobiome and Interkingdom Interactions over the Course of Induction Therapy for Leukemia. mSphere 2020; 5:5/2/e00048-20. [PMID: 32295867 PMCID: PMC7160678 DOI: 10.1128/msphere.00048-20] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Although the term "microbiome" refers to all microorganisms, the majority of microbiome studies focus on the bacteriome. Here, we characterize the oral mycobiome, including mycobiome-bacteriome interactions, in the setting of remission-induction chemotherapy (RIC) for acute myeloid leukemia (AML). Oral samples (n = 299) were prospectively collected twice weekly from 39 AML patients during RIC until neutrophil recovery. Illumina MiSeq 16S rRNA gene (V4) and internal transcribed spacer 2 (ITS2) sequencing were used to determine bacterial and fungal diversity and community composition. Intrakingdom and interkingdom network connectivity at baseline (T1) and at midpoint (T3) and a later time point (T6) were assessed via SPIEC-EASI (sparse inverse covariance estimation for ecological association inference). In this exploratory study, mycobiome α-diversity was not significantly associated with antibiotic or antifungal receipt. However, postchemotherapy mycobiome α-diversity was lower in subjects receiving high-intensity chemotherapy. Additionally, greater decreases in Malassezia levels were seen over time among patients on high-intensity RIC compared to low-intensity RIC (P = 0.003). A significantly higher relative abundance of Candida was found among patients who had infection (P = 0.008), while a significantly higher relative abundance of Fusarium was found among patients who did not get an infection (P = 0.03). Analyses of intrakingdom and interkingdom relationships at T1, T3, and T6 indicated that interkingdom connectivity increased over the course of IC as bacterial α-diversity diminished. In (to our knowledge) the first longitudinal mycobiome study performed during AML RIC, we found that mycobiome-bacteriome interactions are highly dynamic. Our study data suggest that inclusion of mycobiome analysis in the design of microbiome studies may be necessary to optimally understand the ecological and functional role of microbial communities in clinical outcomes.IMPORTANCE This report highlights the importance of longitudinal, parallel characterization of oral fungi and bacteria in order to better elucidate the dynamic changes in microbial community structure and interkingdom functional interactions during the injury of chemotherapy and antibiotic exposure as well as the clinical consequences of these interrelated alterations.
Collapse
|
47
|
Belstrøm D. The salivary microbiota in health and disease. J Oral Microbiol 2020; 12:1723975. [PMID: 32128039 PMCID: PMC7034443 DOI: 10.1080/20002297.2020.1723975] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 12/19/2022] Open
Abstract
The salivary microbiota (SM), comprising bacteria shed from oral surfaces, has been shown to be individualized, temporally stable and influenced by diet and lifestyle. SM reflects local bacterial alterations of the supragingival and subgingival microbiota, and periodontitis and dental-caries associated characteristics of SM have been reported. Also, data suggest an impact of systemic diseases on SM as demonstrated in patients with a wide variety of systemic diseases including diabetes, cancer, HIV and rheumatoid arthritis. The presence of systemic diseases seems to influence salivary levels of specific bacterial species, as well as α- and β-diversity of SM. The composition of SM might thereby potentially mirror oral and general health status. The contentious development of advanced molecular techniques such as metagenomics, metatranscriptomics and metabolomics has enabled the possibility to address bacterial functions rather than presence in microbial samples. However, at present only a few studies have employed such techniques on SM to reveal functional and metabolic characteristics in oral health and disease. Future studies are therefore warranted to illuminate the possible impact of metabolic functions of SM on oral and general health status. Ultimately, such an approach has the possibility to reveal novel and personalized therapeutic avenues in oral and general medicine.
Collapse
Affiliation(s)
- Daniel Belstrøm
- Section for Periodontology and Microbiology, Department of Odontology, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
48
|
Influences of a Prolific Gut Fungus ( Zancudomyces culisetae) on Larval and Adult Mosquito (Aedes aegypti)-Associated Microbiota. Appl Environ Microbiol 2020; 86:AEM.02334-19. [PMID: 31757825 DOI: 10.1128/aem.02334-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 11/12/2019] [Indexed: 11/20/2022] Open
Abstract
Adult mosquitoes inherit a bacterial community from larvae via transstadial transmission, an understudied process that may influence host-microbe interactions. Microbes contribute to important host life history traits, and analyzing transmitted microbial communities, the interrelationship between larval and adult-associated microbiota, and factors influencing host-microbe relationships provides targets for research. During its larval stage, the yellow fever mosquito (Aedes aegypti) hosts the trichomycete gut fungus Zancudomyces culisetae, and fungal colonization coincides with environmental perturbations in the digestive tract microecosystem. Natural populations are differentially exposed to fungi, thereby potentially harboring distinct microbiota and experiencing disparate host-microbe interactions. This study's objectives were to characterize larval and initial adult microbiomes, investigate variation in diversity and distribution of microbial communities across individuals, and assess whether larval fungal colonization impacted microbiomes at these developmental stages. Laboratory-based fungal infestation assays, sequencing of 16S rRNA gene amplicons, and bacterial load quantification protocols revealed that initial adult microbiomes varied in diversity and distribution. Larval fungal colonization had downstream effects on initial adult microbiomes, significantly reducing microbial community variation, shifting relative abundances of certain bacterial families, and influencing transstadial transmission outcomes of particular genera. Further, abundances of several families consistently decreased in adults relative to levels in larvae, possibly reflecting impacts of host development on specific bacterial taxa. These findings demonstrated that a prolific gut fungus impacted mosquito-associated microbiota at two developmental stages in an insect connected with global human health.IMPORTANCE Mosquitoes are widespread vectors of numerous human pathogens and harbor microbiota known to affect host phenotypic traits. However, little research has directly investigated how bacterial communities associated with larvae and adults are connected. We characterized whole-body bacterial communities in mosquito larvae preceding pupation and in newly emerged adults, and investigated whether a significant biotic factor, fungal colonization of the larval hindgut, impacted these microbiomes. Results showed that fungal colonization reduced microbial community variation across individuals and differentially impacted the outcomes of transstadial transmission for certain bacterial genera, revealing downstream effects of the fungus on initial adult microbiomes. The importance of our research is in providing a thorough comparative analysis of whole-body microbiota harbored in larvae and adults of the yellow fever mosquito (Aedes aegypti) and in demonstrating the important role a widespread gut fungus played in a host-associated microbiome.
Collapse
|
49
|
Abstract
OPINION STATEMENT There are approximately 1.2 million new hematologic malignancy cases resulting in ~ 690,000 deaths each year worldwide, and hematologic malignancies remain the most commonly occurring cancer in children. Even though advances in anticancer treatment regimens in recent decades have considerably improved survival rates, their cytotoxic effects and the resulting long-term complications pose a significant burden on the patients and the health care system. Therefore, non-toxic treatment modalities are needed to decrease side effects. The human body is the host to approximately 40 trillion microbes, known as the human microbiota. The large majority of the microbiota is located in the gastrointestinal tract, and is primarily composed of bacteria. The microbiota plays several important physiological roles, ranging from digestive functions to immunological and neural development. Investigating the microbiota in patients with hematologic malignancies has several important implications. The microbiota affects hematopoiesis, and influences the efficacies of chemotherapy and antimicrobial treatments. Determination of the microbiota composition and diversity could be an important part of risk stratification in the future, and may also take part to personalize antimicrobial treatments. Modulation of the microbiota via probiotics or fecal transplant can potentially be involved in reducing side effects of chemotherapy, and eliminating multiple drug resistant strains in patients with hematologic malignancies.
Collapse
|
50
|
Berding K, Donovan SM. Dietary Patterns Impact Temporal Dynamics of Fecal Microbiota Composition in Children With Autism Spectrum Disorder. Front Nutr 2020; 6:193. [PMID: 31998741 PMCID: PMC6968728 DOI: 10.3389/fnut.2019.00193] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 12/13/2019] [Indexed: 12/12/2022] Open
Abstract
Environmental factors such as diet are known influencers on gastrointestinal (GI) microbiota variability and some diseases are associated with microbial stability. Whether microbial variability is related to symptoms of Autism Spectrum Disorder (ASD) and how diet impacts microbial stability in ASD is unknown. Herein, temporal variability in stool microbiota in relation to dietary habits in 2–7 years-old children with ASD (ASD, n = 26) and unaffected controls (CONT, n = 32) was investigated. Fecal samples were collected at baseline, 6-weeks and 6-months. Bacterial composition was assessed using 16S rRNA sequencing. Short fatty acid (SCFA) concentrations were analyzed by gas chromatography. Nutrient intake was assessed using a 3-day food diary and dietary patterns (DP) were empirically derived from a food frequency questionnaire. Social deficit scores (SOCDEF) were assessed using the Pervasive Developmental Disorder Behavior Inventory-Screening Version (PDDBI-SV). GI symptoms were assessed using the GI severity index. Overall, temporal variability in microbial structure, and membership did not differ between the groups. In children with ASD, abundances of Clostridiaceae, Streptophyta, and Clostridiaceae Clostridium, varied significantly, and concentrations of all SCFAs decreased over time. Variability in community membership was negatively correlated with median SOCDEF scores. Additionally, Clostridiales, Lactococcus, Turicibacter, Dorea, and Phascolarctobacterium were components of a more stable microbiota community in children with ASD. DP1, characterized by vegetables, starchy vegetables, legumes, nuts and seeds, fruit, grains, juice and dairy, was associated with changes in species diversity, abundance of Erysipelotricaceae, Clostridiaceae Clostridium, and Oscillospira and concentrations of propionate, butyrate, isobutyrate and isovalerate in children with ASD. DP2 characterized by fried, protein and starchy foods, “Kid's meals,” condiments, and snacks was associated with variations in microbiota structure, abundance of Clostridiaceae Clostridium, and Oscillospira and changes in all SCFA concentrations. However, no association between microbial stability and SOCDEF or GI severity scores were observed. In conclusion, microbiota composition varies over time in children with ASD, might be related to social deficit scores and can be impacted by diet. Future studies investigating the physiological effect of the changes in specific microbial taxa and metabolites are needed to delineate the impact on ASD symptomology.
Collapse
Affiliation(s)
- Kirsten Berding
- Division of Nutritional Sciences, University of Illinois, Urbana, IL, United States
| | - Sharon M Donovan
- Division of Nutritional Sciences, University of Illinois, Urbana, IL, United States.,Department of Food Science and Human Nutrition, University of Illinois, Urbana, IL, United States.,Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, IL, United States
| |
Collapse
|