Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (45)

Search Parameters:
Keywords = microbiota-related carcinogenesis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
33 pages, 1872 KiB  
Review
Exploring the Epidemiologic Burden, Pathogenetic Features, and Clinical Outcomes of Primary Liver Cancer in Patients with Type 2 Diabetes Mellitus (T2DM) and Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD): A Scoping Review
by Mario Romeo, Fiammetta Di Nardo, Carmine Napolitano, Claudio Basile, Carlo Palma, Paolo Vaia, Marcello Dallio and Alessandro Federico
Diabetology 2025, 6(8), 79; https://doi.org/10.3390/diabetology6080079 (registering DOI) - 4 Aug 2025
Abstract
Background/Objectives: Primary liver cancer (PLC), encompassing hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), constitutes a growing global health concern. Metabolic dysfunction-associated Steatotic Liver Disease (MASLD) and Type 2 diabetes mellitus (T2DM) represent a recurrent epidemiological overlap. Individuals with MASLD and T2DM (MASLD-T2DM) are [...] Read more.
Background/Objectives: Primary liver cancer (PLC), encompassing hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), constitutes a growing global health concern. Metabolic dysfunction-associated Steatotic Liver Disease (MASLD) and Type 2 diabetes mellitus (T2DM) represent a recurrent epidemiological overlap. Individuals with MASLD and T2DM (MASLD-T2DM) are at a higher risk of PLC. This scoping review highlights the epidemiological burden, the classic and novel pathogenetic frontiers, and the potential strategies optimizing the management of PLC in MASLD-T2DM. Methods: A systematic search of the PubMed, Medline, and SCOPUS electronic databases was conducted to identify evidence investigating the pathogenetic mechanisms linking MASLD and T2DM to hepatic carcinogenesis, highlighting the most relevant targets and the relatively emerging therapeutic strategies. The search algorithm included in sequence the filter words: “MASLD”, “liver steatosis”, “obesity”, “metabolic syndrome”, “body composition”, “insulin resistance”, “inflammation”, “oxidative stress”, “metabolic dysfunction”, “microbiota”, “glucose”, “immunometabolism”, “trained immunity”. Results: In the MASD-T2DM setting, insulin resistance (IR) and IR-induced mechanisms (including chronic inflammation, insulin/IGF-1 axis dysregulation, and autophagy), simultaneously with the alterations of gut microbiota composition and functioning, represent crucial pathogenetic factors in hepatocarcinogenesis. Besides, the glucose-related metabolic reprogramming emerged as a crucial pathogenetic moment contributing to cancer progression and immune evasion. In this scenario, lifestyle changes, simultaneously with antidiabetic drugs targeting IR-related effects and gut-liver axis, in parallel with novel approaches modulating immunometabolic pathways, represent promising strategies. Conclusions: Metabolic dysfunction, classically featuring MASLD-T2DM, constitutes a continuously expanding global issue, as well as a critical driver in PLC progression, demanding integrated and personalized interventions to reduce the future burden of disease. Full article
Show Figures

Figure 1

29 pages, 1116 KiB  
Review
The Impact of Oncogenic Viruses on Cancer Development: A Narrative Review
by Maria Karoliny da Silva Torres, Gabriel dos Santos Pereira Neto, Izaura Maria Vieira Cayres Vallinoto, Leonardo Oliveira Reis and Antonio Carlos Rosário Vallinoto
Biology 2025, 14(7), 797; https://doi.org/10.3390/biology14070797 - 1 Jul 2025
Viewed by 850
Abstract
Oncogenic viruses are infectious agents that can cause cancer in humans and animals. They are estimated to be responsible for approximately 12% of human cancers worldwide. These viruses trigger a series of mechanisms that allow them to insert their genetic material into host [...] Read more.
Oncogenic viruses are infectious agents that can cause cancer in humans and animals. They are estimated to be responsible for approximately 12% of human cancers worldwide. These viruses trigger a series of mechanisms that allow them to insert their genetic material into host cells, disrupting normal cellular processes and leading to uncontrolled growth and tumor formation. This article reviews the literature on the main oncogenic viruses and reports on newly identified viruses potentially associated with cancer development, addressing the mechanisms of oncogenesis and the types of cancers associated. In addition, the article brings together the evidence for preventive strategies, such as vaccination, and therapeutic advances in combating oncogenic viral infections. This review discusses the role of early detection and treatment in managing virus-related cancers globally. This article reviews current prevention and treatment strategies, including HPV and HBV vaccines and antiviral therapies, and mentions future approaches like immunotherapies and CRISPR/Cas9. Therefore, this article underscores the importance of studying the dynamics of co-infection and the role of human microbiota in viral persistence and carcinogenesis, which opens new possibilities for combination therapies and microbiome-based interventions to slow the progression of viral-related tumors. Full article
(This article belongs to the Special Issue Unraveling the Tumor-Immune Microenvironment Using Transcriptomics)
Show Figures

Figure 1

13 pages, 501 KiB  
Systematic Review
Bioactive Properties of Hazelnut-Derived Products in Colorectal Cancer Prevention: A Systematic Review of Preclinical and Epidemiological Studies
by Giuseppe Mazzola, Mariangela Rondanelli, Federico Buga, Patrizia Riso and Simone Perna
Foods 2025, 14(13), 2154; https://doi.org/10.3390/foods14132154 - 20 Jun 2025
Viewed by 534
Abstract
Background: Colorectal cancer (CRC) is among the leading causes of cancer-related mortality worldwide, with increasing attention being paid to modifiable dietary factors in its prevention. Hazelnut (Corylus avellana L.) represent a nutrient-dense food rich in unsaturated fats, polyphenols, fiber, and phytosterols, [...] Read more.
Background: Colorectal cancer (CRC) is among the leading causes of cancer-related mortality worldwide, with increasing attention being paid to modifiable dietary factors in its prevention. Hazelnut (Corylus avellana L.) represent a nutrient-dense food rich in unsaturated fats, polyphenols, fiber, and phytosterols, with potential anticarcinogenic properties. This systematic review aimed to evaluate the role of hazelnut consumption in the prevention and modulation of CRC risk, with specific focus on experimental, mechanistic, and preclinical evidence. Methods: Following PRISMA guidelines, a systematic search was conducted in PubMed, Google Scholar, and the Cochrane Library for articles published from 2015 onward. Eligible studies included original in vitro and in vivo models, as well as observational studies, evaluating hazelnut or hazelnut-derived products in relation to CRC-related biological, metabolic, or clinical outcomes. Data extraction focused on bioactive composition, experimental models, molecular pathways, and fecal/metabolic markers of carcinogenesis. Results: A total of 11 studies were included after screening 24 records: 8 in vitro investigations, 2 in vivo animal experiments, and 1 epidemiological study. In vitro studies showed that hazelnut derivatives—including fermented hazelnuts and oil-based extracts—exert antiproliferative effects via BAX/BCL-2 modulation, increased caspase-3 activity, and oxidative stress reduction. In vivo studies confirmed improved lipid metabolism, modulation of bile acid composition (notably reduced lithocholic/deoxycholic acid ratio), and enhanced antioxidant defenses. FIBEROX®, a hazelnut skin extract enriched in dietary fiber, demonstrated promising effects on gut microbiota and bile acid detoxification. Conclusions: Hazelnut and their bioactive compounds may aid CRC prevention through multiple molecular and metabolic pathways. Further human studies are needed to confirm these effects and support dietary recommendations. Full article
Show Figures

Figure 1

25 pages, 687 KiB  
Review
Chronotype and Cancer: Emerging Relation Between Chrononutrition and Oncology from Human Studies
by Justyna Godos, Walter Currenti, Raffaele Ferri, Giuseppe Lanza, Filippo Caraci, Evelyn Frias-Toral, Monica Guglielmetti, Cinzia Ferraris, Vivian Lipari, Stefanía Carvajal Altamiranda, Fabio Galvano, Sabrina Castellano and Giuseppe Grosso
Nutrients 2025, 17(3), 529; https://doi.org/10.3390/nu17030529 - 31 Jan 2025
Cited by 6 | Viewed by 3059
Abstract
Fasting–feeding timing is a crucial pattern implicated in the regulation of daily circadian rhythms. The interplay between sleep and meal timing underscores the importance of maintaining circadian alignment in order to avoid creating a metabolic environment conducive to carcinogenesis following the molecular and [...] Read more.
Fasting–feeding timing is a crucial pattern implicated in the regulation of daily circadian rhythms. The interplay between sleep and meal timing underscores the importance of maintaining circadian alignment in order to avoid creating a metabolic environment conducive to carcinogenesis following the molecular and systemic disruption of metabolic performance and immune function. The chronicity of such a condition may support the initiation and progression of cancer through a variety of mechanisms, including increased oxidative stress, immune suppression, and the activation of proliferative signaling pathways. This review aims to summarize current evidence from human studies and provide an overview of the potential mechanisms underscoring the role of chrononutrition (including time-restricted eating) on cancer risk. Current evidence shows that the morning chronotype, suggesting an alignment between physiological circadian rhythms and eating timing, is associated with a lower risk of cancer. Also, early time-restricted eating and prolonged nighttime fasting were also associated with a lower risk of cancer. The current evidence suggests that the chronotype influences cancer risk through cell cycle regulation, the modulation of metabolic pathways and inflammation, and gut microbiota fluctuations. In conclusion, although there are no clear guidelines on this matter, emerging evidence supports the hypothesis that the role of time-related eating (i.e., time/calorie-restricted feeding and intermittent/periodic fasting) could potentially lead to a reduced risk of cancer. Full article
(This article belongs to the Special Issue Associations between Sleep, Nutrition, and Health)
Show Figures

Figure 1

28 pages, 6016 KiB  
Review
Intratumoral Microbiota: Insights from Anatomical, Molecular, and Clinical Perspectives
by Claudia Lombardo, Rosanna Fazio, Marta Sinagra, Giuseppe Gattuso, Federica Longo, Cinzia Lombardo, Mario Salmeri, Guido Nicola Zanghì and Carla Agata Erika Loreto
J. Pers. Med. 2024, 14(11), 1083; https://doi.org/10.3390/jpm14111083 - 31 Oct 2024
Cited by 4 | Viewed by 2737
Abstract
The human microbiota represents a heterogeneous microbial community composed of several commensal, symbiotic, and even pathogenic microorganisms colonizing both the external and internal body surfaces. Despite the term “microbiota” being commonly used to identify microorganisms inhabiting the gut, several pieces of evidence suggest [...] Read more.
The human microbiota represents a heterogeneous microbial community composed of several commensal, symbiotic, and even pathogenic microorganisms colonizing both the external and internal body surfaces. Despite the term “microbiota” being commonly used to identify microorganisms inhabiting the gut, several pieces of evidence suggest the presence of different microbiota physiologically colonizing other organs. In this context, several studies have also confirmed that microbes are integral components of tumor tissue in different types of cancer, constituting the so-called “intratumoral microbiota”. The intratumoral microbiota is closely related to the occurrence and development of cancer as well as to the efficacy of anticancer treatments. Indeed, intratumoral microbiota can contribute to carcinogenesis and metastasis formation as some microbes can directly cause DNA damage, while others can induce the activation of proinflammatory responses or oncogenic pathways and alter the tumor microenvironment (TME). All these characteristics make the intratumoral microbiota an interesting topic to investigate for both diagnostic and prognostic purposes in order to improve the management of cancer patients. This review aims to gather the most recent data on the role of the intratumoral microbiota in cancer development, progression, and response to treatment, as well as its potential diagnostic and prognostic value. Full article
(This article belongs to the Special Issue Novel Biomarkers in the Diagnostics of Cancer)
Show Figures

Figure 1

32 pages, 2112 KiB  
Review
Antioxidant Role of Probiotics in Inflammation-Induced Colorectal Cancer
by Sevag Hamamah, Andrei Lobiuc and Mihai Covasa
Int. J. Mol. Sci. 2024, 25(16), 9026; https://doi.org/10.3390/ijms25169026 - 20 Aug 2024
Cited by 7 | Viewed by 3545
Abstract
Colorectal cancer (CRC) continues to be a significant contributor to global morbidity and mortality. Emerging evidence indicates that disturbances in gut microbial composition, the formation of reactive oxygen species (ROS), and the resulting inflammation can lead to DNA damage, driving the pathogenesis and [...] Read more.
Colorectal cancer (CRC) continues to be a significant contributor to global morbidity and mortality. Emerging evidence indicates that disturbances in gut microbial composition, the formation of reactive oxygen species (ROS), and the resulting inflammation can lead to DNA damage, driving the pathogenesis and progression of CRC. Notably, bacterial metabolites can either protect against or contribute to oxidative stress by modulating the activity of antioxidant enzymes and influencing signaling pathways that govern ROS-induced inflammation. Additionally, microbiota byproducts, when supplemented through probiotics, can affect tumor microenvironments to enhance treatment efficacy and selectively mediate the ROS-induced destruction of CRC cells. This review aims to discuss the mechanisms by which taxonomical shifts in gut microbiota and related metabolites such as short-chain fatty acids, secondary bile acids, and trimethylamine-N-oxide influence ROS concentrations to safeguard or promote the onset of inflammation-mediated CRC. Additionally, we focus on the role of probiotic species in modulating ROS-mediated signaling pathways that influence both oxidative status and inflammation, such as Nrf2-Keap1, NF-κB, and NLRP3 to mitigate carcinogenesis. Overall, a deeper understanding of the role of gut microbiota on oxidative stress may aid in delaying or preventing the onset of CRC and offer new avenues for adjunct, CRC-specific therapeutic interventions such as cancer immunotherapy. Full article
(This article belongs to the Special Issue Effects of Bioactive Compounds in Oxidative Stress and Inflammation)
Show Figures

Figure 1

24 pages, 1189 KiB  
Review
Role of Gut Microbial Metabolites in the Pathogenesis of Primary Liver Cancers
by Maria Pallozzi, Valeria De Gaetano, Natalia Di Tommaso, Lucia Cerrito, Francesco Santopaolo, Leonardo Stella, Antonio Gasbarrini and Francesca Romana Ponziani
Nutrients 2024, 16(14), 2372; https://doi.org/10.3390/nu16142372 - 22 Jul 2024
Cited by 9 | Viewed by 3663
Abstract
Hepatobiliary malignancies, which include hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are the sixth most common cancers and the third leading cause of cancer-related death worldwide. Hepatic carcinogenesis is highly stimulated by chronic inflammation, defined as fibrosis deposition, and an aberrant imbalance between liver [...] Read more.
Hepatobiliary malignancies, which include hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), are the sixth most common cancers and the third leading cause of cancer-related death worldwide. Hepatic carcinogenesis is highly stimulated by chronic inflammation, defined as fibrosis deposition, and an aberrant imbalance between liver necrosis and nodular regeneration. In this context, the gut–liver axis and gut microbiota have demonstrated a critical role in the pathogenesis of HCC, as dysbiosis and altered intestinal permeability promote bacterial translocation, leading to chronic liver inflammation and tumorigenesis through several pathways. A few data exist on the role of the gut microbiota or bacteria resident in the biliary tract in the pathogenesis of CCA, and some microbial metabolites, such as choline and bile acids, seem to show an association. In this review, we analyze the impact of the gut microbiota and its metabolites on HCC and CCA development and the role of gut dysbiosis as a biomarker of hepatobiliary cancer risk and of response during anti-tumor therapy. We also discuss the future application of gut microbiota in hepatobiliary cancer management. Full article
(This article belongs to the Special Issue Diet, Gut Microbiota, and Gastrointestinal Disease)
Show Figures

Figure 1

25 pages, 744 KiB  
Review
Understanding the Conundrum of Pancreatic Cancer in the Omics Sciences Era
by Alberto Nicoletti, Mattia Paratore, Federica Vitale, Marcantonio Negri, Giuseppe Quero, Giorgio Esposto, Irene Mignini, Sergio Alfieri, Antonio Gasbarrini, Maria Assunta Zocco and Lorenzo Zileri Dal Verme
Int. J. Mol. Sci. 2024, 25(14), 7623; https://doi.org/10.3390/ijms25147623 - 11 Jul 2024
Cited by 5 | Viewed by 2768
Abstract
Pancreatic cancer (PC) is an increasing cause of cancer-related death, with a dismal prognosis caused by its aggressive biology, the lack of clinical symptoms in the early phases of the disease, and the inefficacy of treatments. PC is characterized by a complex tumor [...] Read more.
Pancreatic cancer (PC) is an increasing cause of cancer-related death, with a dismal prognosis caused by its aggressive biology, the lack of clinical symptoms in the early phases of the disease, and the inefficacy of treatments. PC is characterized by a complex tumor microenvironment. The interaction of its cellular components plays a crucial role in tumor development and progression, contributing to the alteration of metabolism and cellular hyperproliferation, as well as to metastatic evolution and abnormal tumor-associated immunity. Furthermore, in response to intrinsic oncogenic alterations and the influence of the tumor microenvironment, cancer cells undergo a complex oncogene-directed metabolic reprogramming that includes changes in glucose utilization, lipid and amino acid metabolism, redox balance, and activation of recycling and scavenging pathways. The advent of omics sciences is revolutionizing the comprehension of the pathogenetic conundrum of pancreatic carcinogenesis. In particular, metabolomics and genomics has led to a more precise classification of PC into subtypes that show different biological behaviors and responses to treatments. The identification of molecular targets through the pharmacogenomic approach may help to personalize treatments. Novel specific biomarkers have been discovered using proteomics and metabolomics analyses. Radiomics allows for an earlier diagnosis through the computational analysis of imaging. However, the complexity, high expertise required, and costs of the omics approach are the main limitations for its use in clinical practice at present. In addition, the studies of extracellular vesicles (EVs), the use of organoids, the understanding of host–microbiota interactions, and more recently the advent of artificial intelligence are helping to make further steps towards precision and personalized medicine. This present review summarizes the main evidence for the application of omics sciences to the study of PC and the identification of future perspectives. Full article
(This article belongs to the Special Issue Molecular Research on the Pancreatic Cancer)
Show Figures

Figure 1

15 pages, 4051 KiB  
Article
Effect of Trace Element Selenium on the Intestinal Microbial Community in Nude Mice with Colorectal Cancer
by Yintong Su, Xiaohua Cai, Xingxing Fan, Jiayu Ning and Mei Shen
Microorganisms 2024, 12(7), 1336; https://doi.org/10.3390/microorganisms12071336 - 29 Jun 2024
Viewed by 1663
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. The role of intestinal microbiota in carcinogenesis has also become an important research topic, and CRC is closely related to the intestinal microbiota. Selenium-containing compounds have attracted more attention as anticancer drugs as [...] Read more.
Colorectal cancer (CRC) is the third most common cancer worldwide. The role of intestinal microbiota in carcinogenesis has also become an important research topic, and CRC is closely related to the intestinal microbiota. Selenium-containing compounds have attracted more attention as anticancer drugs as they can have minimal side effects. The purpose of this study was to determine and compare the effect of sodium selenite and selenomethionine on the microbial communities of nude mice with CRC. A CRC ectopic tumorigenesis model was established by subcutaneously injecting HCT116 cells into nude mice. The mice were then intraperitoneally injected with sodium selenite and selenomethionine for 24 days to regulate their intestinal microbiota. Compared with sodium selenite, selenomethionine resulted in a greater reduction in the richness and diversity of intestinal microbiota in nude mice with CRC, and the richness and diversity were closer to healthy levels. Selenomethionine also regulated a wider variety of flora. Additionally, sodium selenite and selenomethionine produced different microorganisms, changed function and metabolic pathways in the intestinal microbiota. Both sodium selenite and selenomethionine have certain effects on restoring the intestinal microbial diversity in nude mice with CRC, and the effect of selenomethionine is better than that of sodium selenite. Full article
(This article belongs to the Section Medical Microbiology)
Show Figures

Figure 1

15 pages, 705 KiB  
Review
Exploring the Link between Helicobacter pylori, Gastric Microbiota and Gastric Cancer
by Costantino Sgamato, Alba Rocco, Debora Compare, Kateryna Priadko, Marco Romano and Gerardo Nardone
Antibiotics 2024, 13(6), 484; https://doi.org/10.3390/antibiotics13060484 - 24 May 2024
Cited by 10 | Viewed by 3178
Abstract
Gastric cancer (GC) still represents one of the leading causes of cancer-related mortality and is a major public health issue worldwide. Understanding the etiopathogenetic mechanisms behind GC development holds immense potential to revolutionize patients’ treatment and prognosis. Within the complex web of genetic [...] Read more.
Gastric cancer (GC) still represents one of the leading causes of cancer-related mortality and is a major public health issue worldwide. Understanding the etiopathogenetic mechanisms behind GC development holds immense potential to revolutionize patients’ treatment and prognosis. Within the complex web of genetic predispositions and environmental factors, the connection between Helicobacter pylori (H. pylori) and gastric microbiota emerges as a focus of intense research investigation. According to the most recent hypotheses, H. pylori triggers inflammatory responses and molecular alterations in gastric mucosa, while non-Helicobacter microbiota modulates disease progression. In this review, we analyze the current state of the literature on the relationship between H. pylori and non-Helicobacter gastric microbiota in gastric carcinogenesis, highlighting the mechanisms by which microecological dysbiosis can contribute to the malignant transformation of the mucosa. Full article
(This article belongs to the Special Issue Pathogenesis, Diagnosis and Treatment of H. pylori Infection)
Show Figures

Figure 1

11 pages, 231 KiB  
Systematic Review
The Use of Fecal Microbiota Transplant in Overcoming and Modulating Resistance to Anti-PD-1 Therapy in Patients with Skin Cancer
by Tahne Vongsavath, Rodd Rahmani, Kyaw Min Tun and Vignan Manne
Cancers 2024, 16(3), 499; https://doi.org/10.3390/cancers16030499 - 24 Jan 2024
Cited by 5 | Viewed by 2818
Abstract
While immune checkpoint inhibitors have evolved into the standard of care for advanced melanoma, 40–50% of melanoma cases progress while on therapies. The relationship between bacterium and carcinogenesis is well founded, such as in H. pylori in gastric cancers, and Fusobacterium in colorectal [...] Read more.
While immune checkpoint inhibitors have evolved into the standard of care for advanced melanoma, 40–50% of melanoma cases progress while on therapies. The relationship between bacterium and carcinogenesis is well founded, such as in H. pylori in gastric cancers, and Fusobacterium in colorectal cancers. This interplay between dysbiosis and carcinogenesis questions whether changes in the microbiome could affect treatment. Thus, FMT may find utility in modifying the efficacy of anti-PD-1. This review aims to examine the use of FMT in treatment-resistant melanoma. A literature search was performed using the keywords “fecal microbiota transplant” and “skin cancer”. Studies were reviewed for inclusion criteria and quality and in the final stage, and three studies were included. Overall objective responses were reported in 65% of patients who were able to achieve CR, and 45% who achieved PR. Clinical benefit rate of combined CR/PR with stable disease greater or equal to 6 months was 75%. Reported objective responses found durable stable disease lasting 12 months. Overall survival was 7 months, and overall PRS was 3 months. As for the evaluation of safety, many patients reported grade 1–2 FMT related AE. Only following the administration of anti-PD-1 therapy were there a grade 3 or higher AE. Full article
13 pages, 661 KiB  
Review
The Microbiome in the Obesity-Breast Cancer Axis: Diagnostic and Therapeutic Potential
by Dimiter Avtanski, Varun Reddy, Radoslav Stojchevski, Nikola Hadzi-Petrushev and Mitko Mladenov
Pathogens 2023, 12(12), 1402; https://doi.org/10.3390/pathogens12121402 - 29 Nov 2023
Cited by 9 | Viewed by 2819
Abstract
A growing body of evidence has demonstrated a relationship between the microbiome, adiposity, and cancer development. The microbiome is emerging as an important factor in metabolic disease and cancer pathogenesis. This review aimed to highlight the role of the microbiome in obesity and [...] Read more.
A growing body of evidence has demonstrated a relationship between the microbiome, adiposity, and cancer development. The microbiome is emerging as an important factor in metabolic disease and cancer pathogenesis. This review aimed to highlight the role of the microbiome in obesity and its association with cancer, with a particular focus on breast cancer. This review discusses how microbiota dysbiosis may contribute to obesity and obesity-related diseases, which are linked to breast cancer. It also explores the potential of the gut microbiome to influence systemic immunity, leading to carcinogenesis via the modulation of immune function. This review underscores the potential use of the microbiome profile as a diagnostic tool and treatment target, with strategies including probiotics, fecal microbiota transplantation, and dietary interventions. However, this emphasizes the need for more research to fully understand the complex relationship between the microbiome, metabolic disorders, and breast cancer. Future studies should focus on elucidating the mechanisms underlying the impact of the microbiome on breast cancer and exploring the potential of the microbiota profile as a biomarker and treatment target. Full article
(This article belongs to the Special Issue Role of Microorganisms in Breast Cancer)
Show Figures

Figure 1

19 pages, 2871 KiB  
Review
Role of Ubiquitination and Epigenetics in the Regulation of AhR Signaling in Carcinogenesis and Metastasis: “Albatross around the Neck” or “Blessing in Disguise”
by Ammad Ahmad Farooqi, Venera Rakhmetova, Gulnara Kapanova, Gulnur Tanbayeva, Akmaral Mussakhanova, Akmaral Abdykulova and Alma-Gul Ryskulova
Cells 2023, 12(19), 2382; https://doi.org/10.3390/cells12192382 - 29 Sep 2023
Cited by 3 | Viewed by 2953
Abstract
The molecular mechanisms and signal transduction cascades evoked by the activation of aryl hydrocarbon receptor (AhR) are becoming increasingly understandable. AhR is a ligand-activated transcriptional factor that integrates environmental, dietary and metabolic cues for the pleiotropic regulation of a wide variety of mechanisms. [...] Read more.
The molecular mechanisms and signal transduction cascades evoked by the activation of aryl hydrocarbon receptor (AhR) are becoming increasingly understandable. AhR is a ligand-activated transcriptional factor that integrates environmental, dietary and metabolic cues for the pleiotropic regulation of a wide variety of mechanisms. AhR mediates transcriptional programming in a ligand-specific, context-specific and cell-type-specific manner. Pioneering cutting-edge research works have provided fascinating new insights into the mechanistic role of AhR-driven downstream signaling in a wide variety of cancers. AhR ligands derived from food, environmental contaminants and intestinal microbiota strategically activated AhR signaling and regulated multiple stages of cancer. Although AhR has classically been viewed and characterized as a ligand-regulated transcriptional factor, its role as a ubiquitin ligase is fascinating. Accordingly, recent evidence has paradigmatically shifted our understanding and urged researchers to drill down deep into these novel and clinically valuable facets of AhR biology. Our rapidly increasing realization related to AhR-mediated regulation of the ubiquitination and proteasomal degradation of different proteins has started to scratch the surface of intriguing mechanisms. Furthermore, AhR and epigenome dynamics have shown previously unprecedented complexity during multiple stages of cancer progression. AhR not only transcriptionally regulated epigenetic-associated molecules, but also worked with epigenetic-modifying enzymes during cancer progression. In this review, we have summarized the findings obtained not only from cell-culture studies, but also from animal models. Different clinical trials are currently being conducted using AhR inhibitors and PD-1 inhibitors (Pembrolizumab and nivolumab), which confirm the linchpin role of AhR-related mechanistic details in cancer progression. Therefore, further studies are required to develop a better comprehension of the many-sided and “diametrically opposed” roles of AhR in the regulation of carcinogenesis and metastatic spread of cancer cells to the secondary organs. Full article
Show Figures

Figure 1

17 pages, 1363 KiB  
Review
Bioactive Compounds of Dietary Origin and Their Influence on Colorectal Cancer as Chemoprevention
by Paulina Delgado-Gonzalez, Elsa N. Garza-Treviño, David A. de la Garza Kalife, Adriana Quiroz Reyes and Esther Alhelí Hernández-Tobías
Life 2023, 13(10), 1977; https://doi.org/10.3390/life13101977 - 28 Sep 2023
Cited by 9 | Viewed by 3171
Abstract
Colorectal cancer (CRC) is one of the most common causes of death and the third most diagnosed cancer worldwide. The tumor microenvironment and cancer stem cells participate in colorectal tumor progression and can dictate malignancy. Nutrition status affects treatment response and the progression [...] Read more.
Colorectal cancer (CRC) is one of the most common causes of death and the third most diagnosed cancer worldwide. The tumor microenvironment and cancer stem cells participate in colorectal tumor progression and can dictate malignancy. Nutrition status affects treatment response and the progression or recurrence of the tumor. This review summarizes the main bioactive compounds against the molecular pathways related to colorectal carcinogenesis. Moreover, we focus on the compounds with chemopreventive properties, mainly polyphenols and carotenoids, which are highly studied dietary bioactive compounds present in major types of food, like vegetables, fruits, and seeds. Their proprieties are antioxidant and gut microbiota modulation, important in the intestine because they decrease reactive oxygen species and inflammation, both principal causes of cancer. These compounds can promote apoptosis and inhibit cell growth, proliferation, and migration. Combined with oncologic treatment, a sensitization to first-line colorectal chemotherapy schemes, such as FOLFOX and FOLFIRI, is observed, making them an attractive and natural support in the oncologic treatment of CRC. Full article
(This article belongs to the Section Epidemiology)
Show Figures

Figure 1

22 pages, 1338 KiB  
Review
Role of the Microbiota in Skin Neoplasms: New Therapeutic Horizons
by Paola Savoia, Barbara Azzimonti, Roberta Rolla and Elisa Zavattaro
Microorganisms 2023, 11(10), 2386; https://doi.org/10.3390/microorganisms11102386 - 25 Sep 2023
Cited by 8 | Viewed by 3324
Abstract
The skin and the gut are regularly colonized by a variety of microorganisms capable of interacting with the immune system through their metabolites and influencing the balance between immune tolerance and inflammation. Alterations in the composition and diversity of the skin microbiota have [...] Read more.
The skin and the gut are regularly colonized by a variety of microorganisms capable of interacting with the immune system through their metabolites and influencing the balance between immune tolerance and inflammation. Alterations in the composition and diversity of the skin microbiota have been described in various cutaneous diseases, including skin cancer, and the actual function of the human microbiota in skin carcinogenesis, such as in progression and metastasis, is currently an active area of research. The role of Human Papilloma Virus (HPV) in the pathogenesis of squamous cell carcinoma is well consolidated, especially in chronically immunosuppressed patients. Furthermore, an imbalance between Staphylococcus spp., such as Staphylococcus epidermidis and aureus, has been found to be strongly related to the progression from actinic keratosis to squamous cell carcinoma and differently associated with various stages of the diseases in cutaneous T-cell lymphoma patients. Also, in melanoma patients, differences in microbiota have been related to dissimilar disease course and prognosis and may affect the effectiveness and tolerability of immune checkpoint inhibitors, which currently represent one of the best chances of a cure. From this point of view, acting on microbiota can be considered a possible therapeutic option for patients with advanced skin cancers, even if several issues are still open. Full article
(This article belongs to the Special Issue Bacteria and Fungi Probiotics)
Show Figures

Figure 1

Back to TopTop