Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
Mol Microbiol ; 2024 May 01.
Article in English | MEDLINE | ID: mdl-38690771

ABSTRACT

The small intestine represents a complex and understudied gut niche with significant implications for human health. Indeed, many infectious and non-infectious diseases center within the small intestine and present similar clinical manifestations to large intestinal disease, complicating non-invasive diagnosis and treatment. One major neglected aspect of small intestinal diseases is the feedback relationship with the resident collection of commensal organisms, the gut microbiota. Studies focused on microbiota-host interactions in the small intestine in the context of infectious and non-infectious diseases are required to identify potential therapeutic targets dissimilar from those used for large bowel diseases. While sparsely populated, the small intestine represents a stringent commensal bacterial microenvironment the host relies upon for nutrient acquisition and protection against invading pathogens (colonization resistance). Indeed, recent evidence suggests that disruptions to host-microbiota interactions in the small intestine impact enteric bacterial pathogenesis and susceptibility to non-infectious enteric diseases. In this review, we focus on the microbiota's impact on small intestine function and the pathogenesis of infectious and non-infectious diseases of the gastrointestinal (GI) tract. We also discuss gaps in knowledge on the role of commensal microorganisms in proximal GI tract function during health and disease.

2.
mBio ; 15(2): e0269223, 2024 Feb 14.
Article in English | MEDLINE | ID: mdl-38226819

ABSTRACT

A recent study published in mBio by Nemet et al. revealed the critical role played by two gut microbiota members in producing the metabolites indoxyl sulfate (IS) and p-cresol sulfate (pCS) (I. Nemet, M. Funabashi,X. S. Li, M. Dwidar, et al., 2023, mBio 14:e01331-23, https://doi.org/10.1128/mbio.01331-23). Understanding microbial pathways leading to IS and pCS production is crucial because they are connected to a pre-thrombotic profile, and having high levels of these metabolites increases the risk of cardiovascular diseases (CVD). Hence, this study can offer vital insights into assessing the risk for CVD and identifying potential treatment targets for this disease.


Subject(s)
Cardiovascular Diseases , Cresols , Microbiota , Sulfuric Acid Esters , Thrombosis , Humans , Indican
3.
bioRxiv ; 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37961401

ABSTRACT

Salmonella Typhimurium is an enteric pathogen that employs a variety of mechanisms to exploit inflammation resulting in expansion in the intestinal tract, but host factors that contribute to or counteract the luminal expansion are not well-defined. Endoplasmic reticulum (ER) stress induces inflammation and plays an important role in the pathogenesis of infectious diseases. However, little is known about the contribution of ER stress-induced inflammation during Salmonella pathogenesis. Here, we demonstrate that the ER stress markers Hspa5 and Xbp1 are induced in the colon of S. Typhimurium infected mice, but the pro-apoptotic transcription factor Ddit3, that encodes for the protein CHOP, is significantly downregulated. S. Typhimurium-infected mice deficient for CHOP displayed a significant decrease in inflammation, colonization, dissemination, and pathology compared to littermate control mice. Preceding the differences in S. Typhimurium colonization, a significant decrease in Nos2 gene and iNOS protein expression was observed. Deletion of Chop decreased the bioavailability of nitrate in the colon leading to reduced fitness advantage of wild type S. Typhimurium over a napA narZ narG mutant strain (deficient in nitrate respiration). CD11b+ myeloid cells, but not intestinal epithelial cells, produced iNOS resulting in nitrate bioavailability for S. Typhimurium to expand in the intestinal tract in a CHOP-dependent manner. Altogether our work demonstrates that the host protein CHOP facilitates iNOS expression in CD11b+ cells thereby contributing to luminal expansion of S. Typhimurium via nitrate respiration.

4.
Cell Host Microbe ; 31(10): 1604-1619.e10, 2023 10 11.
Article in English | MEDLINE | ID: mdl-37794592

ABSTRACT

The mechanisms by which the early-life microbiota protects against environmental factors that promote childhood obesity remain largely unknown. Using a mouse model in which young mice are simultaneously exposed to antibiotics and a high-fat (HF) diet, we show that Lactobacillus species, predominant members of the small intestine (SI) microbiota, regulate intestinal epithelial cells (IECs) to limit diet-induced obesity during early life. A Lactobacillus-derived metabolite, phenyllactic acid (PLA), protects against metabolic dysfunction caused by early-life exposure to antibiotics and a HF diet by increasing the abundance of peroxisome proliferator-activated receptor γ (PPAR-γ) in SI IECs. Therefore, PLA is a microbiota-derived metabolite that activates protective pathways in the small intestinal epithelium to regulate intestinal lipid metabolism and prevent antibiotic-associated obesity during early life.


Subject(s)
Microbiota , Pediatric Obesity , Humans , Child , Animals , Mice , Lipid Metabolism , Diet, High-Fat/adverse effects , Anti-Bacterial Agents , Polyesters , Mice, Inbred C57BL
5.
Infect Immun ; 91(11): e0032223, 2023 Nov 16.
Article in English | MEDLINE | ID: mdl-37800916

ABSTRACT

One of the major contributors to child mortality in the world is diarrheal diseases, with an estimated 800,000 deaths per year. Many pathogens are causative agents of these illnesses, including the enteropathogenic or enterohemorrhagic forms of Escherichia coli. These bacteria are characterized by their ability to cause attaching and effacing lesions in the gut mucosa. Although much has been learned about the pathogenicity of these organisms and the immune response against them, the role of the intestinal microbiota during these infections is not well characterized. Infection of mice with E. coli requires pre-treatment with antibiotics in most mouse models, which hinders the study of the microbiota in an undisturbed environment. Using Citrobacter rodentium as a murine model for attaching and effacing bacteria, we show that C57BL/6 mice deficient in granzyme B expression are highly susceptible to severe disease caused by C. rodentium infection. Although a previous publication from our group shows that granzyme B-deficient CD4+ T cells are partially responsible for this phenotype, in this report, we present data demonstrating that the microbiota, in particular members of the order Turicibacterales, have an important role in conferring resistance. Mice deficient in Turicibacter sanguinis have increased susceptibility to severe disease. However, when these mice are co-housed with resistant mice or colonized with T. sanguinis, susceptibility to severe infection is reduced. These results clearly suggest a critical role for this commensal in the protection against enteropathogens.


Subject(s)
Enterobacteriaceae Infections , Escherichia coli , Child , Humans , Animals , Mice , Citrobacter rodentium/genetics , Granzymes , Enterobacteriaceae Infections/microbiology , Mice, Inbred C57BL , Bacteria
6.
Cell Host Microbe ; 31(10): 1639-1654.e10, 2023 10 11.
Article in English | MEDLINE | ID: mdl-37776864

ABSTRACT

During intestinal inflammation, host nutritional immunity starves microbes of essential micronutrients, such as iron. Pathogens scavenge iron using siderophores, including enterobactin; however, this strategy is counteracted by host protein lipocalin-2, which sequesters iron-laden enterobactin. Although this iron competition occurs in the presence of gut bacteria, the roles of commensals in nutritional immunity involving iron remain unexplored. Here, we report that the gut commensal Bacteroides thetaiotaomicron acquires iron and sustains its resilience in the inflamed gut by utilizing siderophores produced by other bacteria, including Salmonella, via a secreted siderophore-binding lipoprotein XusB. Notably, XusB-bound enterobactin is less accessible to host sequestration by lipocalin-2 but can be "re-acquired" by Salmonella, allowing the pathogen to evade nutritional immunity. Because the host and pathogen have been the focus of studies of nutritional immunity, this work adds commensal iron metabolism as a previously unrecognized mechanism modulating the host-pathogen interactions and nutritional immunity.


Subject(s)
Salmonella Infections , Siderophores , Humans , Lipocalin-2/metabolism , Siderophores/metabolism , Enterobactin/metabolism , Bacteria/metabolism , Iron/metabolism
7.
bioRxiv ; 2023 Jun 26.
Article in English | MEDLINE | ID: mdl-37425782

ABSTRACT

During intestinal inflammation, host nutritional immunity starves microbes of essential micronutrients such as iron. Pathogens scavenge iron using siderophores, which is counteracted by the host using lipocalin-2, a protein that sequesters iron-laden siderophores, including enterobactin. Although the host and pathogens compete for iron in the presence of gut commensal bacteria, the roles of commensals in nutritional immunity involving iron remain unexplored. Here, we report that the gut commensal Bacteroides thetaiotaomicron acquires iron in the inflamed gut by utilizing siderophores produced by other bacteria including Salmonella, via a secreted siderophore-binding lipoprotein termed XusB. Notably, XusB-bound siderophores are less accessible to host sequestration by lipocalin-2 but can be "re-acquired" by Salmonella , allowing the pathogen to evade nutritional immunity. As the host and pathogen have been the focus of studies of nutritional immunity, this work adds commensal iron metabolism as a previously unrecognized mechanism modulating the interactions between pathogen and host nutritional immunity.

8.
bioRxiv ; 2023 Apr 25.
Article in English | MEDLINE | ID: mdl-37163036

ABSTRACT

One of the major contributors to child mortality in the world is diarrheal diseases, with an estimated 800,000 deaths per year. Many pathogens are causative agents of these illnesses, including the enteropathogenic (EPEC) or enterohemorrhagic (EHEC) forms of Escherichia coli. These bacteria are characterized by their ability to cause attaching and effacing lesions in the gut mucosa. Although much has been learned about the pathogenicity of these organisms and the immune response against them, the role of the intestinal microbiota during these infections is not well characterized. Infection of mice with E. coli requires pre-treatment with antibiotics in most mouse models, which hinders the study of the microbiota in an undisturbed environment. Using Citrobacter rodentium as a murine model for attaching and effacing bacteria, we show that C57BL/6 mice deficient in granzyme B expression are highly susceptible to severe disease caused by C. rodentium infection. Although a previous publication from our group shows that granzyme B-deficient CD4+ T cells are partially responsible for this phenotype, in this report we present data demonstrating that the microbiota, in particular members of the order Turicibacterales, have an important role in conferring resistance. Mice deficient in Turicibacter sanguinis have increased susceptibility to severe disease. However, when these mice are co-housed with resistant mice, or colonized with T. sanguinis, susceptibility to severe infection is reduced. These results clearly suggest a critical role for this commensal in the protection against entero-pathogens.

9.
Gut Microbes ; 15(1): 2185029, 2023.
Article in English | MEDLINE | ID: mdl-36872510

ABSTRACT

The mouse cecum has emerged as a model system for studying microbe-host interactions, immunoregulatory functions of the microbiome, and metabolic contributions of gut bacteria. Too often, the cecum is falsely considered as a uniform organ with an evenly distributed epithelium. We developed the cecum axis (CecAx) preservation method to show gradients in epithelial tissue architecture and cell types along the cecal ampulla-apex and mesentery-antimesentery axes. We used imaging mass spectrometry of metabolites and lipids to suggest functional differences along these axes. Using a model of Clostridioides difficile infection, we show how edema and inflammation are unequally concentrated along the mesenteric border. Finally, we show the similarly increased edema at the mesenteric border in two models of Salmonella enterica serovar Typhimurium infection as well as enrichment of goblet cells along the antimesenteric border. Our approach facilitates mouse cecum modeling with detailed attention to inherent structural and functional differences within this dynamic organ.


Subject(s)
Gastrointestinal Microbiome , Animals , Mice , Cecum , Epithelium , Goblet Cells , Host Microbial Interactions
10.
Cell Rep ; 42(2): 112128, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36807140

ABSTRACT

The cytokine interleukin-23 (IL-23) is involved in the pathogenesis of inflammatory and autoimmune conditions including inflammatory bowel disease (IBD). IL23R is enriched in intestinal Tregs, yet whether IL-23 modulates intestinal Tregs remains unknown. Here, investigating IL-23R signaling in Tregs specifically, we show that colonic Tregs highly express Il23r compared with Tregs from other compartments and their frequency is reduced upon IL-23 administration and impairs Treg suppressive function. Similarly, colonic Treg frequency is increased in mice lacking Il23r specifically in Tregs and exhibits a competitive advantage over IL-23R-sufficient Tregs during inflammation. Finally, IL-23 antagonizes liver X receptor pathway, cellular cholesterol transporter Abca1, and increases Treg apoptosis. Our results show that IL-23R signaling regulates intestinal Tregs by increasing cell turnover, antagonizing suppression, and decreasing cholesterol efflux. These results suggest that IL-23 negatively regulates Tregs in the intestine with potential implications for promoting chronic inflammation in patients with IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Humans , Mice , Colitis/pathology , Forkhead Transcription Factors/metabolism , Inflammation/pathology , Inflammatory Bowel Diseases/pathology , Interleukin-23/metabolism , T-Lymphocytes, Regulatory
11.
Cell Mol Gastroenterol Hepatol ; 14(4): 731-750, 2022.
Article in English | MEDLINE | ID: mdl-35835390

ABSTRACT

BACKGROUND & AIMS: Inflammatory bowel disease (IBD) is characterized by severe gastrointestinal inflammation, but many patients experience extra-intestinal disease. Bone loss is one common extra-intestinal manifestation of IBD that occurs through dysregulated interactions between osteoclasts and osteoblasts. Systemic inflammation has been postulated to contribute to bone loss, but the specific pathologic mechanisms have not yet been fully elucidated. We hypothesized that intestinal inflammation leads to bone loss through increased abundance and altered function of osteoclast progenitors. METHODS: We used chemical, T cell driven, and infectious models of intestinal inflammation to determine the impact of intestinal inflammation on bone volume, the skeletal cytokine environment, and the cellular changes to pre-osteoclast populations within bone marrow. Additionally, we evaluated the potential for monoclonal antibody treatment against an inflammation-induced osteoclast co-receptor, myeloid DNAX activation protein 12-associating lectin-1 (MDL-1) to reduce bone loss during colitis. RESULTS: We observed significant bone loss across all models of intestinal inflammation. Bone loss was associated with an increase in pro-osteoclastogenic cytokines within the bone and an expansion of a specific Cd11b-/loLy6Chi osteoclast precursor (OCP) population. Intestinal inflammation led to altered OCP expression of surface receptors involved in osteoclast differentiation and function, including the pro-osteoclastogenic co-receptor MDL-1. OCPs isolated from mice with intestinal inflammation demonstrated enhanced osteoclast differentiation ex vivo compared to controls, which was abrogated by anti-MDL-1 antibody treatment. Importantly, in vivo anti-MDL-1 antibody treatment ameliorated bone loss during intestinal inflammation. CONCLUSIONS: Collectively, these data implicate the pathologic expansion and altered function of OCPs expressing MDL-1 in bone loss during IBD.


Subject(s)
Bone Resorption , Inflammatory Bowel Diseases , Lectins, C-Type , Osteoclasts , Osteogenesis , Receptors, Cell Surface , Animals , Antibodies, Monoclonal/metabolism , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation/physiology , Cytokines/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestines/metabolism , Lectins/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis/genetics , Osteogenesis/physiology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
12.
Cell Host Microbe ; 30(6): 836-847.e6, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35568027

ABSTRACT

Changes in the microbiota composition are associated with many human diseases, but factors that govern strain abundance remain poorly defined. We show that a commensal Escherichia coli strain and a pathogenic Salmonella enterica serovar Typhimurium isolate both utilize nitrate for intestinal growth, but each accesses this resource in a distinct biogeographical niche. Commensal E. coli utilizes epithelial-derived nitrate, whereas nitrate in the niche occupied by S. Typhimurium is derived from phagocytic infiltrates. Surprisingly, avirulent S. Typhimurium was shown to be unable to utilize epithelial-derived nitrate because its chemotaxis receptors McpB and McpC exclude the pathogen from the niche occupied by E. coli. In contrast, E. coli invades the niche constructed by S. Typhimurium virulence factors and confers colonization resistance by competing for nitrate. Thus, nutrient niches are not defined solely by critical resources, but they can be further subdivided biogeographically within the host into distinct microhabitats, thereby generating new niche opportunities for distinct bacterial species.


Subject(s)
Gastrointestinal Microbiome , Salmonella typhimurium , Escherichia coli , Humans , Nitrates , Nutrients
13.
Infect Immun ; 90(3): e0001322, 2022 03 17.
Article in English | MEDLINE | ID: mdl-35100011

ABSTRACT

Research on Brucella pathogenesis has focused primarily on its ability to cause persistent intracellular infection of the mononuclear phagocyte system. At these sites, Brucella abortus evades innate immunity, which results in low-level inflammation and chronic infection of phagocytes. In contrast, the host response in the placenta during infection is characterized by severe inflammation and extensive extracellular replication of B. abortus. Despite the importance of reproductive disease caused by Brucella infection, our knowledge of the mechanisms involved in placental inflammation and abortion is limited. To understand the immune responses specifically driving placental pathology, we modeled placental B. abortus infection in pregnant mice. B. abortus infection caused an increase in the production of tumor necrosis factor alpha (TNF-α), specifically in the placenta. We found that placental expression levels of Tnfa and circulating TNF-α were dependent on the induction of endoplasmic reticulum stress and the B. abortus type IV secretion system (T4SS) effector protein VceC. Blockade of TNF-α reduced placental inflammation and improved fetal viability in mice. This work sheds light on a tissue-specific response of the placenta to B. abortus infection that may be important for bacterial transmission via abortion in the natural host species.


Subject(s)
Brucellosis, Bovine , Brucellosis , Animals , Brucella abortus/physiology , Brucellosis/microbiology , Cattle , Female , Inflammation , Mice , Placenta , Pregnancy , Tumor Necrosis Factor-alpha/metabolism
14.
Lancet Oncol ; 23(3): e116-e128, 2022 03.
Article in English | MEDLINE | ID: mdl-35090673

ABSTRACT

Sporadic colorectal cancer has traditionally been viewed as a malignancy of older individuals. However, as the global prevalence of the disease diagnosed in younger individuals (<50 years) is expected to increase within the next decade, greater recognition is now being given to early-onset colorectal cancer. The cause of the predicted rise in prevalence is largely unknown and probably multifactorial. In this Series paper, we discuss the potential underlying causes of early-onset colorectal cancer, the role of energy balance, biological and genomic mechanisms (including microbiome aspects), and the treatment of early-onset colorectal cancer. We have specifically considered the psychosocial challenges of being diagnosed with colorectal cancer at younger age and the potential financial toxicity that might ensue. This Series paper brings a comprehensive review based on the existing data in the hopes of optimising the overall outcomes for patients with early-onset colorectal cancer.


Subject(s)
Colorectal Neoplasms , Age of Onset , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , Genomics , Humans , Prevalence
15.
Cell Rep ; 38(1): 110180, 2022 01 04.
Article in English | MEDLINE | ID: mdl-34986344

ABSTRACT

The gut microbiota benefits the host by limiting enteric pathogen expansion (colonization resistance), partially via the production of inhibitory metabolites. Propionate, a short-chain fatty acid produced by microbiota members, is proposed to mediate colonization resistance against Salmonella enterica serovar Typhimurium (S. Tm). Here, we show that S. Tm overcomes the inhibitory effects of propionate by using it as a carbon source for anaerobic respiration. We determine that propionate metabolism provides an inflammation-dependent colonization advantage to S. Tm during infection. Such benefit is abolished in the intestinal lumen of Salmonella-infected germ-free mice. Interestingly, S. Tm propionate-mediated intestinal expansion is restored when germ-free mice are monocolonized with Bacteroides thetaiotaomicron (B. theta), a prominent propionate producer in the gut, but not when mice are monocolonized with a propionate-production-deficient B. theta strain. Taken together, our results reveal a strategy used by S. Tm to mitigate colonization resistance by metabolizing microbiota-derived propionate.


Subject(s)
Anaerobiosis/physiology , Propionates/metabolism , Salmonella Infections, Animal/pathology , Salmonella typhimurium/growth & development , Salmonella typhimurium/metabolism , Animals , Antibiosis/physiology , Bacteroides thetaiotaomicron/genetics , Bacteroides thetaiotaomicron/metabolism , Female , Gastrointestinal Microbiome/physiology , Germ-Free Life , Intestines/microbiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Knockout , Nitrates/metabolism
16.
Cell Host Microbe ; 29(11): 1606-1608, 2021 11 10.
Article in English | MEDLINE | ID: mdl-34762826

ABSTRACT

Have you ever caught family members eating the last piece of your Halloween candy? In this issue of Cell Host & Microbe, Osbelt et al. and Eberl et al. demonstrate how commensal Enterobacteriaceae preempt pathogen carbohydrate utilization, dependent upon the composition of the surrounding gut microbial community.


Subject(s)
Enterobacteriaceae , Microbiota , Candy , Carbohydrates , Symbiosis
17.
Curr Opin Microbiol ; 64: 82-90, 2021 12.
Article in English | MEDLINE | ID: mdl-34688039

ABSTRACT

The intestine is home to a large and complex bacterial ecosystem (microbiota), which performs multiple beneficial functions for the host, including immune education, nutrition, and protection against invasion by enteric pathogens (colonization resistance). The host and microbiome symbiotic interactions occur in part through metabolic crosstalk. Thus, microbiota members have evolved highly diverse metabolic pathways to inhibit pathogen colonization via activation of protective immune responses and nutrient acquisition and utilization. Conversely, pathogenic Enterobacteriaceae actively induce an inflammation-dependent disruption of the gut microbial ecosystem (dysbiosis) to gain a competitive metabolic advantage against the resident microbiota. This review discusses the recent findings on the crucial role of microbiota metabolites in colonization resistance regulation. Additionally, we summarize metabolic mechanisms used by pathogenic Enterobacteriaceae to outcompete commensal microbes and cause disease.


Subject(s)
Enterobacteriaceae Infections , Gastrointestinal Microbiome , Dysbiosis , Ecosystem , Enterobacteriaceae/genetics , Humans
18.
Science ; 373(6556): 813-818, 2021 08 13.
Article in English | MEDLINE | ID: mdl-34385401

ABSTRACT

A Western-style, high-fat diet promotes cardiovascular disease, in part because it is rich in choline, which is converted to trimethylamine (TMA) by the gut microbiota. However, whether diet-induced changes in intestinal physiology can alter the metabolic capacity of the microbiota remains unknown. Using a mouse model of diet-induced obesity, we show that chronic exposure to a high-fat diet escalates Escherichia coli choline catabolism by altering intestinal epithelial physiology. A high-fat diet impaired the bioenergetics of mitochondria in the colonic epithelium to increase the luminal bioavailability of oxygen and nitrate, thereby intensifying respiration-dependent choline catabolism of E. coli In turn, E. coli choline catabolism increased levels of circulating trimethlamine N-oxide, which is a potentially harmful metabolite generated by gut microbiota.


Subject(s)
Colon/physiology , Diet, High-Fat , Escherichia coli/metabolism , Intestinal Mucosa/physiology , Methylamines/metabolism , Animals , Cell Hypoxia , Choline/administration & dosage , Choline/metabolism , Colon/cytology , Energy Metabolism , Epithelial Cells/physiology , Escherichia coli/genetics , Escherichia coli/growth & development , Feces/microbiology , Gastrointestinal Microbiome , Inflammation , Intestinal Mucosa/metabolism , Male , Methylamines/blood , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Nitrates/metabolism , Obesity , Oxygen Consumption
19.
Pathog Dis ; 79(6)2021 07 28.
Article in English | MEDLINE | ID: mdl-34283236

ABSTRACT

Mentoring is a developmental experience intended to increase the willingness to learn and establish credibility while building positive relationships through networking. In this commentary, we focus on intentional mentoring for underrepresented mentees, including individuals that belong to minority racial, ethnic and gender identity groups in Science, Technology, Engineering, Mathematics and Medicine (STEMM) fields. Intentional mentoring is the superpower action necessary for developing harmony and comprehending the purpose and value of the mentor/mentee relationship. Regardless of a mentor's career stage, we believe the strategies discussed may be used to create a supportive and constructive mentorship environment; thereby improving the retention rates of underrepresented mentees within the scientific community.


Subject(s)
Communication , Interprofessional Relations , Mentoring , Mentors , Humans , Trust
20.
Dis Model Mech ; 14(5)2021 05 01.
Article in English | MEDLINE | ID: mdl-34060626

ABSTRACT

Landmark discoveries in the gut microbiome field have paved the way for new research aimed at illuminating the influence of microbiota in colorectal cancer. A major challenge is to account for the effect of inherently variable environmental factors on the host and the gut microbiome, while concurrently determining their contribution to carcinogenesis. Here, we briefly discuss the role of the gut microbial community in colorectal cancer and elaborate on the recent insight that environmental factors related to a Western diet and lifestyle may drive the bloom of tumorigenic members of the gut microbiota. We also discuss how future research focused on untangling host-microbe interactions in the colon may influence medical insights that relate to the prevention and treatment of colorectal cancer.


Subject(s)
Colorectal Neoplasms/complications , Dysbiosis/complications , Life Style , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/therapy , Dysbiosis/microbiology , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Host-Pathogen Interactions , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...