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

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (185)

Search Parameters:
Keywords = ProCRC

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 9009 KiB  
Article
Cancer-Associated Fibroblasts Establish Spatially Distinct Prognostic Niches in Subcutaneous Colorectal Cancer Mouse Model
by Zhixian Lin, Jinmeng Wang, Yixin Ma, Yanan Zhu, Yuhan Li, Zhengtao Xiao and Wei Zhao
Cancers 2025, 17(14), 2402; https://doi.org/10.3390/cancers17142402 - 19 Jul 2025
Viewed by 323
Abstract
Background/Objectives: Subcutaneous tumor models are widely used in colorectal cancer (CRC) research due to their experimental accessibility; however, the spatial organization and regulatory mechanisms of their tumor microenvironment remain poorly understood. Methods: Here, we applied spatial transcriptomics to systematically characterize spatial heterogeneity within [...] Read more.
Background/Objectives: Subcutaneous tumor models are widely used in colorectal cancer (CRC) research due to their experimental accessibility; however, the spatial organization and regulatory mechanisms of their tumor microenvironment remain poorly understood. Methods: Here, we applied spatial transcriptomics to systematically characterize spatial heterogeneity within MC38 subcutaneous tumors in a syngeneic mouse model. Results: We identified two spatially distinct tumor zones, partitioned by cancer-associated fibroblasts (CAFs), that differ markedly in cellular composition, oncogenic signaling, immune infiltration, and metabolic states. One zone exhibited features of TGF-β-driven extracellular matrix remodeling, immune exclusion, and hyperproliferative metabolism, while the other was enriched for immunosuppressive macrophages, metabolic reprogramming via PPAR and AMPK pathways, and high-risk cell populations. Spatially resolved cell–cell communication networks further revealed zone-specific ligand–receptor interactions—such as ANGPTL4–SDC2 and PROS1–AXL—that underpin stromal remodeling and immune evasion and are associated with patient prognosis. Conclusions: Collectively, our study uncovers how region-specific cellular ecosystems and intercellular crosstalk establish prognostically divergent niches within subcutaneous CRC tumors, offering insights into spatially guided therapeutic strategies. Full article
(This article belongs to the Section Tumor Microenvironment)
Show Figures

Figure 1

16 pages, 663 KiB  
Article
A Disintegrin and Metalloprotease 15 (ADAM15) as a Potential Predictor of Distant Metastasis in Colorectal Cancer (CRC)
by Adrianna Romanowicz, Marta Łukaszewicz-Zając, Barbara Choromańska, Sara Pączek, Hady Razak Hady, Piotr Myśliwiec, Jacek Jamiołkowski, Piotr Stępniewski, Leszek Kozłowski and Barbara Mroczko
J. Clin. Med. 2025, 14(14), 5082; https://doi.org/10.3390/jcm14145082 - 17 Jul 2025
Viewed by 216
Abstract
Background: The pro-tumorigenic role of a disintegrin and metalloprotease 15 (ADAM15) is supported by its modified expression in primary tumors and ability to promote tumor growth in colorectal cancer (CRC). Cancer cell-derived ADAM15 promotes the progression of this malignancy by modulating the tumor [...] Read more.
Background: The pro-tumorigenic role of a disintegrin and metalloprotease 15 (ADAM15) is supported by its modified expression in primary tumors and ability to promote tumor growth in colorectal cancer (CRC). Cancer cell-derived ADAM15 promotes the progression of this malignancy by modulating the tumor microenvironment. However, according to our knowledge, this study is the first to assess serum ADAM15 concentrations in CRC patients in comparison to classical tumor markers—carcinoembryonic antigen (CEA) and cancer antigen 19-9 (CA19-9)—and a marker of the inflammatory process, C-reactive protein (CRP). The aim was to evaluate whether circulating serum ADAM15 might be a candidate biomarker for CRC diagnosis and progression. Methods: The study included 110 CRC patients and 54 healthy volunteers. Serum concentrations of ADAM15, CEA, and CA19-9 were measured using immunoenzyme assays, while CRP levels were assessed by the turbidimetric method. Diagnostic characteristics of all tested proteins were calculated. Results: Serum ADAM15 and classical tumor marker (CEA and CA19) levels were higher in CRC patients than in healthy subjects. However, a significant difference was observed only for CEA (p < 0.001). ADAM15 concentrations were significantly higher in CRC patients with distant metastases compared to those without metastases (p = 0.043). The highest diagnostic sensitivity (89%) was achieved by combined analysis of ADAM15 and CRP levels. Conclusions: These findings suggest a significant role of ADAM15 in CRC pathogenesis, indicating the usefulness of this protein in the prediction of distant metastases. Measurement of serum ADAM15, especially in combination with classical tumor markers (CEA) and inflammation markers (CRP), may improve the diagnosis of patients with CRC. Full article
(This article belongs to the Section General Surgery)
Show Figures

Figure 1

28 pages, 1513 KiB  
Review
The Impact of the Microbiota on the Immune Response Modulation in Colorectal Cancer
by Ana Iulia Neagu, Marinela Bostan, Vlad Alexandru Ionescu, Gina Gheorghe, Camelia Mia Hotnog, Viviana Roman, Mirela Mihaila, Simona Isabelle Stoica, Camelia Cristina Diaconu, Carmen Cristina Diaconu, Simona Maria Ruta and Coralia Bleotu
Biomolecules 2025, 15(7), 1005; https://doi.org/10.3390/biom15071005 - 14 Jul 2025
Viewed by 455
Abstract
Colorectal cancer (CRC) is a multifactorial disease increasingly recognized for its complex interplay with the gut microbiota. The disruption of microbial homeostasis—dysbiosis—has profound implications for intestinal barrier integrity and host immune function. Pathogenic bacterial species such as Fusobacterium nucleatum, Escherichia coli harboring polyketide [...] Read more.
Colorectal cancer (CRC) is a multifactorial disease increasingly recognized for its complex interplay with the gut microbiota. The disruption of microbial homeostasis—dysbiosis—has profound implications for intestinal barrier integrity and host immune function. Pathogenic bacterial species such as Fusobacterium nucleatum, Escherichia coli harboring polyketide synthase (pks) island, and enterotoxigenic Bacteroides fragilis are implicated in CRC through mechanisms involving mucosal inflammation, epithelial barrier disruption, and immune evasion. These pathogens promote pro-tumorigenic inflammation, enhance DNA damage, and suppress effective anti-tumor immunity. Conversely, commensal and probiotic bacteria, notably Lactobacillus and Bifidobacterium species, exert protective effects by preserving epithelial barrier function and priming host immune responses. These beneficial microbes can promote the maturation of dendritic cells, stimulate CD8+ T cell cytotoxicity, and modulate regulatory T cell populations, thereby enhancing anti-tumor immunity. The dichotomous role of the microbiota underscores its potential as both a biomarker and a therapeutic target in CRC. Recent advances in studies have explored microbiota-modulating strategies—ranging from dietary interventions and prebiotics to fecal microbiota transplantation (FMT) and microbial consortia—as adjuncts to conventional therapies. Moreover, the composition of the gut microbiome has been shown to influence the responses to immunotherapy and chemotherapy, raising the possibility of microbiome-informed precision oncology therapy. This review synthesizes the current findings on the pathogenic and protective roles of bacteria in CRC and evaluates the translational potential of microbiome-based interventions in shaping future therapeutic paradigms. Full article
Show Figures

Figure 1

26 pages, 6233 KiB  
Review
Colonic Aging and Colorectal Cancer: An Unignorable Interplay and Its Translational Implications
by Qiyan Yin, Fen Qin, Fangliu Gan, Guangxi Zhao, Ronghua Chen, Yue Wen, Xueyang Hua, Fugui Zeng, Yuezheng Zhang, Yuliang Xiao, Wenbing Xie and Yong Tao
Biology 2025, 14(7), 805; https://doi.org/10.3390/biology14070805 - 3 Jul 2025
Viewed by 443
Abstract
Colorectal cancer (CRC) incidence increases markedly with age, yet chronological age is an inadequate proxy for the complex biological processes involved. Colon aging, the intrinsic biological aging of the colonic tissue, is emerging as a crucial, active driver of CRC development. This review [...] Read more.
Colorectal cancer (CRC) incidence increases markedly with age, yet chronological age is an inadequate proxy for the complex biological processes involved. Colon aging, the intrinsic biological aging of the colonic tissue, is emerging as a crucial, active driver of CRC development. This review comprehensively analyzes the interplay between colon aging and CRC pathogenesis by examining fundamental hallmarks of aging—such as altered tissue homeostasis, epigenetic dysregulation, and microenvironmental shifts including chronic inflammation (inflammaging), gut microbiome dysbiosis, and extracellular matrix remodeling—manifest specifically within the aging colon to synergistically foster a pro-tumorigenic environment. Key findings synthesized from the literature highlight the critical roles of impaired colonic stem cell function, epithelial barrier disruption (“leaky gut”), persistent low-grade inflammation, and altered microbial communities and their metabolites in promoting CRC initiation and progression. Translating this mechanistic understanding holds significant promise: insights from colon aging research can inform novel biomarkers for improved early detection and risk stratification, guide the development of personalized preventative strategies and therapeutic interventions targeting aging pathways, and underpin public health initiatives focused on healthy colon aging. Ultimately, recognizing colon aging as a modifiable contributor to CRC offers a powerful avenue to potentially reduce CRC incidence and enhance patient outcomes, particularly in the vulnerable aging population. Full article
(This article belongs to the Section Cancer Biology)
Show Figures

Figure 1

21 pages, 3190 KiB  
Article
Pyrvinium Pamoate and BCL-XL Inhibitors Act Synergistically to Kill Patient-Derived Colorectal Adenoma Organoids
by Maree C. Faux, Chenkai Ma, Serena R. Kane, Andre Samson, Yumiko Hirokawa, Ilka Priebe, Leah Cosgrove, Rajvinder Singh, Michael Christie, Gregor Brown, Kim Y. C. Fung and Antony W. Burgess
Organoids 2025, 4(3), 15; https://doi.org/10.3390/organoids4030015 - 2 Jul 2025
Viewed by 281
Abstract
Current systemic therapies for advanced colorectal cancer (CRC) have limited efficacy, so more effective strategies for the treatment and prevention of CRC are needed. The majority of colorectal cancers are initiated by mutations in Wnt signalling pathway genes, including mutations in the APC [...] Read more.
Current systemic therapies for advanced colorectal cancer (CRC) have limited efficacy, so more effective strategies for the treatment and prevention of CRC are needed. The majority of colorectal cancers are initiated by mutations in Wnt signalling pathway genes, including mutations in the APC gene, which result in a truncated APC protein and lead to excess signalling from β-catenin and the formation of pre-cancerous adenomas. The aim of this study was to determine if targeting the Wnt pathway in combination with pro-apoptotic mimetics altered the proliferative capacity or viability of human colorectal adenoma cells. Patient-derived colorectal adenoma organoid cultures were established from colon adenoma tissue collected by colonoscopy and recapitulated the histopathology of primary colorectal adenoma tissue. The growth of colorectal adenoma organoids is inhibited by the Wnt-signalling antagonist pyrvinium pamoate (PP) and a pro-apoptotic inhibitor of BCL-XL but not BCL-2 (venetoclax) or MCL-1 inhibitors. At low concentrations, the PP and the BCL-XL inhibitor combination demonstrated potent synergy and induced apoptosis in APC-defective patient-derived adenoma organoids, even in the presence of oncogenic KRAS or BRAF mutations, providing a new strategy for colon cancer prevention. Full article
Show Figures

Figure 1

22 pages, 8293 KiB  
Article
Time-Dependent Impact of Betulin and Its Derivatives on IL-8 Expression in Colorectal Cancer Cells with Molecular Docking Studies
by Marcel Madej, Adrianna Halama, Elwira Chrobak and Joanna Magdalena Gola
Int. J. Mol. Sci. 2025, 26(13), 6186; https://doi.org/10.3390/ijms26136186 - 27 Jun 2025
Viewed by 322
Abstract
Colorectal cancer (CRC) remains one of the most prevalent malignancies of the gastrointestinal tract worldwide, with chronic inflammation recognized as a key factor in its progression. Among pro-inflammatory cytokines, interleukin 8 (IL-8) plays a pivotal role in promoting angiogenesis, tumor cell migration, and [...] Read more.
Colorectal cancer (CRC) remains one of the most prevalent malignancies of the gastrointestinal tract worldwide, with chronic inflammation recognized as a key factor in its progression. Among pro-inflammatory cytokines, interleukin 8 (IL-8) plays a pivotal role in promoting angiogenesis, tumor cell migration, and metastasis. Elevated IL-8 expression is frequently associated with advanced CRC stages. This study investigated the effects of betulin and its semi-synthetic derivatives, EB5 and ECH147, on IL-8 expression in CRC cell lines characterized by differing malignancy grades. IL-8 transcript and protein levels were quantified using real-time RT-qPCR and a proximity ligation assay, respectively, following compound exposure at 2, 8, and 24 h. Basal IL-8 levels were significantly higher in low-grade CRC cell lines. Among the compounds tested, ECH147 exerted the most pronounced, time-dependent inhibitory effect on CXCL8 expression. Furthermore, molecular docking analyses revealed that ECH147 exhibits stronger binding affinity toward the IL-8 protein compared to conventional chemotherapeutics. These findings suggest that the modification of the betulin structure via the incorporation of a propynoyl moiety enhances both its molecular interaction with CXCL8 and its anti-inflammatory potential. ECH147 and EB5 thus emerge as promising candidates for further development as immunomodulatory agents targeting the IL-8-associated pathway in CRC. Full article
(This article belongs to the Special Issue Natural Products in Cancer Prevention and Treatment)
Show Figures

Figure 1

21 pages, 1422 KiB  
Review
Cannabidiol (CBD) and Colorectal Tumorigenesis: Potential Dual Modulatory Roles via the Serotonergic Pathway
by Zhenhua Liu
Curr. Oncol. 2025, 32(7), 375; https://doi.org/10.3390/curroncol32070375 - 26 Jun 2025
Viewed by 547
Abstract
The 2018 Farm Bill legalized hemp-derived cannabidiol (CBD) products containing less than 0.3% tetrahydrocannabinol (THC) in the United States. This legislative shift catalyzed both public and scientific interest in CBD’s potential health benefits. However, the rapid expansion of the CBD market has considerably [...] Read more.
The 2018 Farm Bill legalized hemp-derived cannabidiol (CBD) products containing less than 0.3% tetrahydrocannabinol (THC) in the United States. This legislative shift catalyzed both public and scientific interest in CBD’s potential health benefits. However, the rapid expansion of the CBD market has considerably outpaced rigorous scientific research, leaving many health claims largely unsubstantiated. While preclinical studies suggest that CBD may exert antitumorigenic effects in colorectal cancer (CRC) by modulating cell proliferation, apoptosis, and inflammation, clinical evidence supporting these effects remains limited. This review critically examines the current evidence on the role of CBD in colorectal tumorigenesis, with particular attention to its molecular mechanisms and interactions with the serotonergic system—a signaling pathway implicated in the development of CRC and possessing potential dual anti- and pro-tumorigenic properties. By influencing the serotonergic system, CBD may confer both protective and potentially deleterious effects during CRC development. This review underscores the need for further research to elucidate the complex mechanisms of CBD in colorectal tumorigenesis and to evaluate its therapeutic potential in clinical settings. Understanding these interactions could pave the way for novel prevention and treatment strategies, optimizing the anticancer efficacy of CBD while mitigating unintended risks. Full article
Show Figures

Figure 1

27 pages, 2282 KiB  
Review
The Anti-Inflammatory, Immunomodulatory, and Pro-Autophagy Activities of Probiotics for Colorectal Cancer Prevention and Treatment: A Narrative Review
by Beatrice Garavaglia, Letizia Vallino, Alessandra Ferraresi, Annalisa Visciglia, Angela Amoruso, Marco Pane, Camelia Munteanu and Ciro Isidoro
Biomedicines 2025, 13(7), 1554; https://doi.org/10.3390/biomedicines13071554 - 25 Jun 2025
Viewed by 476
Abstract
Colorectal cancer (CRC) is a major global health concern, particularly in Western countries where there is high consumption of processed food. Gut microbiota, intestinal inflammation, and autophagy play pivotal roles in CRC initiation and progression. Probiotics and probiotic metabolites (particularly short-chain fatty acids) [...] Read more.
Colorectal cancer (CRC) is a major global health concern, particularly in Western countries where there is high consumption of processed food. Gut microbiota, intestinal inflammation, and autophagy play pivotal roles in CRC initiation and progression. Probiotics and probiotic metabolites (particularly short-chain fatty acids) have emerged as potential preventive and adjuvant therapeutics by restoring a balanced gut microbiota, dampening inflammation, stimulating immune response, and improving barrier integrity and intestinal epithelial homeostasis by modulating autophagy. This narrative review discusses the current evidence supporting the anti-inflammatory, immunomodulatory, and pro-autophagy effects of probiotics and their metabolites and explores their potential preventive and therapeutic applications in CRC management. Full article
Show Figures

Graphical abstract

29 pages, 1900 KiB  
Article
MSC1 Cells Suppress Colorectal Cancer Cell Growth via Metabolic Reprogramming, Laminin–Integrin Adhesion Signaling, Oxidative Stress Resistance, and a Tumor-Suppressive Secretome
by Panagiota-Angeliki Galliou, Niti Argyri, Papaioannou Maria, George Koliakos and Nikolaos A. Papanikolaou
Biomedicines 2025, 13(6), 1503; https://doi.org/10.3390/biomedicines13061503 - 19 Jun 2025
Viewed by 633
Abstract
Background/Objectives: Mesenchymal stem cells (MSCs) possess immunomodulatory properties, tumor-homing, and low immunogenicity, making them attractive for cell-based cancer therapies, but their role in colorectal cancer (CRC) remains controversial. The MSC1 phenotype, a pro-inflammatory, tumor-suppressive state induced by short-term, low-dose LPS activation via TLR4, [...] Read more.
Background/Objectives: Mesenchymal stem cells (MSCs) possess immunomodulatory properties, tumor-homing, and low immunogenicity, making them attractive for cell-based cancer therapies, but their role in colorectal cancer (CRC) remains controversial. The MSC1 phenotype, a pro-inflammatory, tumor-suppressive state induced by short-term, low-dose LPS activation via TLR4, has shown therapeutic promise but remains poorly characterized in CRC. We aimed to elucidate MSC1’s tumor-suppressive mechanisms and validate its activity against CRC cells using an integrated bioinformatics and in vitro approach. Methods: We constructed a high-confidence protein-protein interaction (PPI) network in Wharton’s jelly-derived MSCs (WJ-MSCs) following TLR4 activation to uncover enriched signaling pathways, transcriptional regulators, and secreted factors. Functional and transcriptional enrichment analyses pinpointed key mechanisms. We then co-cultured MSC1 cells with CRC cells to assess effects on proliferation and metabolism. Results: Network analysis revealed six tumor-suppressive mechanisms of MSC1 cells: (i) Metabolic reprogramming via enhanced glucose and lipid uptake, phosphoinositide signaling, and membrane/protein recycling, (ii) Robust antioxidant defenses, including SOS signaling and system xc⁻, (iii) Extracellular matrix stabilization and laminin-111–integrin-mediated adhesion, (iv) Secretome with direct anti-cancer effects, (v) Regulation of survival and cancer-associated fibroblasts (CAFs) formation inhibition through balanced proliferation, apoptosis, and epigenetic signals, (vi) Controlled pro-inflammatory signaling with anti-inflammatory feedback. In vitro, MSC1 cells significantly suppressed CRC cell proliferation and metabolic activity versus controls. Conclusions: This study provides the first mechanistic map of MSC1’s tumor-suppressive functions in CRC, extending beyond immunomodulation to include metabolic competition, ECM stabilization, and anti-cancer secretome activity. These findings establish MSC1 cells as a novel therapeutic strategy for CRC in cell-based cancer therapies. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Graphical abstract

17 pages, 44923 KiB  
Article
Inhibition of PCSK9 Attenuates Liver Endothelial Cell Activation Induced by Colorectal Cancer Stem Cells During Liver Metastasis
by Ander Martin, Daniela Gerovska, Marcos J. Arauzo-Bravo, Maitane Duarte García-Escudero, Helena García García, Iratxe Bañares, Naroa Fontal, Geraldine Siegfried, Serge Evrad, Simon Pernot, Abdel-Majid Khatib and Iker Badiola
Cancers 2025, 17(12), 1977; https://doi.org/10.3390/cancers17121977 - 13 Jun 2025
Viewed by 690
Abstract
Background: Colorectal cancer (CRC) is among the most prevalent and lethal cancers globally, with liver metastasis representing the leading cause of CRC-related mortality. Proprotein convertase subtilisin/kexin type 9 (PCSK9) has recently gained attention due to its overexpression in colorectal tumor tissues and its [...] Read more.
Background: Colorectal cancer (CRC) is among the most prevalent and lethal cancers globally, with liver metastasis representing the leading cause of CRC-related mortality. Proprotein convertase subtilisin/kexin type 9 (PCSK9) has recently gained attention due to its overexpression in colorectal tumor tissues and its potential role in driving metastatic progression. This aims to investigate the involvement of PCSK9 in the liver metastatic niche, focusing on its effects on liver sinusoidal endothelial cells (LSECs), key components of the liver microenvironment. Methods: LSECs were stimulated with conditioned media derived from differentiated colorectal cancer cells and cancer stem cells (CSCs), the latter generated by reprogramming SW620 and CT26 cell lines. RNA sequencing was used to profile gene expression in LSECs. PCSK9 mRNA and protein levels were quantified by qPCR and Western blotting, respectively. PCSK9 expression in CRC liver metastases was evaluated by immunofluorescent staining. Results: PCSK9 was detected in both human and murine LSECs and significantly upregulated following exposure to CSC-conditioned media. Immunofluorescent staining confirmed PCSK9 expression in LSECs within CRC liver metastases. Total RNA sequencing revealed that a pre-treatment of LSECs with the PCSK9 inhibitor PF-06446864 prior to CSC stimulation seems to reduce the expression of microRNAs linked to cell migration and proliferation. Functional assays demonstrated that CSC-conditioned media enhanced LSEC proliferation and migration, effects reversed by PCSK9 inhibition. Conclusions: PCSK9 promotes the activation of LSECs in response to colorectal CSCs, contributing to a pro-metastatic phenotype. These findings highlight PCSK9 as a potential therapeutic target in colorectal liver metastasis. Full article
Show Figures

Figure 1

55 pages, 2579 KiB  
Review
Regulation and Function of Tumor-Associated Macrophages (TAMs) in Colorectal Cancer (CRC): The Role of the SRIF System in Macrophage Regulation
by Agnieszka Geltz, Jakub Geltz and Aldona Kasprzak
Int. J. Mol. Sci. 2025, 26(11), 5336; https://doi.org/10.3390/ijms26115336 - 1 Jun 2025
Viewed by 1220
Abstract
Colorectal cancer (CRC) remains the leading cause of morbidity and mortality for both men and women worldwide. Tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME) of solid tumors, including CRC. These macrophages are found in the pro-inflammatory [...] Read more.
Colorectal cancer (CRC) remains the leading cause of morbidity and mortality for both men and women worldwide. Tumor-associated macrophages (TAMs) are the most abundant immune cells in the tumor microenvironment (TME) of solid tumors, including CRC. These macrophages are found in the pro-inflammatory M1 and anti-inflammatory M2 forms, with the latter increasingly recognized for its tumor-promoting phenotypes. Many signaling molecules and pathways, including AMPK, EGFR, STAT3/6, mTOR, NF-κB, MAPK/ERK, and HIFs, are involved in regulating TAM polarization. Consequently, researchers are investigating several potential predictive and prognostic markers, and novel TAM-based therapeutic targets, especially in combination therapies for CRC. Macrophages of the gastrointestinal tract, including the normal colon and rectum, produce growth hormone-releasing inhibitory peptide/somatostatin (SRIF/SST) and five SST receptors (SSTRs, SST1-5). While the immunosuppressive function of the SRIF system is primarily known for various tissues, its role within CRC-associated TAMs remains underexplored. This review focuses on the following three aspects of TAMs: first, the role of macrophages in the normal colon and rectum within the broader context of macrophage biology; second, the various bioactive factors and signaling pathways associated with TAM function, along with potential strategies targeting TAMs in CRC; and third, the interaction between the SRIF system and macrophages in both normal tissues and the CRC microenvironment. Full article
(This article belongs to the Special Issue The Role of Macrophages in Cancers)
Show Figures

Figure 1

8 pages, 907 KiB  
Proceeding Paper
Unsaturated 10H2DA Queen Bee Acid from Royal Jelly Modulates Epithelial-to-Mesenchymal Transition in SW-480 Colorectal Cancer Cells
by Milena M. Jovanović and Dragana S. Šeklić
Biol. Life Sci. Forum 2025, 43(1), 3; https://doi.org/10.3390/blsf2025043003 - 26 May 2025
Viewed by 359
Abstract
Cancer research largely focuses on epithelial-to-mesenchymal transition (EMT) as a critical mechanism required for the formation of metastases. This process involves the transformation of epithelial cells into mesenchymal cells by acquiring suppressed levels of anti-EMT and elevated expression of pro-EMT markers. Unsaturated fatty [...] Read more.
Cancer research largely focuses on epithelial-to-mesenchymal transition (EMT) as a critical mechanism required for the formation of metastases. This process involves the transformation of epithelial cells into mesenchymal cells by acquiring suppressed levels of anti-EMT and elevated expression of pro-EMT markers. Unsaturated fatty acid 10H2DA has not been investigated hitherto regarding its potential to target specific EMT markers in colorectal cancer (CRC). In our study, this substance showed successful upregulation of the expression of the anti-EMT marker E-cadherin and downregulation of the expression of pro-EMT markers SNAIL, N-cadherin, and Vimentin at the gene and protein levels. This prominent effect of 10H2DA in modulating the expression of specific and significant EMT markers in CRC should not be neglected in future studies regarding anticancer therapeutic approaches. Full article
(This article belongs to the Proceedings of The 2nd International Electronic Conference on Genes)
Show Figures

Figure 1

13 pages, 4323 KiB  
Article
D-Limonene Exhibits Antiproliferative Activity Against Human Colorectal Adenocarcinoma (Caco-2) Cells via Regulation of Inflammatory and Apoptotic Pathways
by Abdullah A. A. Alghamdi
Curr. Issues Mol. Biol. 2025, 47(5), 370; https://doi.org/10.3390/cimb47050370 - 18 May 2025
Viewed by 617
Abstract
Current therapies for colorectal cancer (CRC) are associated with significant side effects and limitations, driving the search for novel therapeutic approaches. This study investigated the antiproliferative potential of D-limonene, a natural compound, on human colorectal adenocarcinoma (Caco-2) cells and analyzed its underlying mechanisms. [...] Read more.
Current therapies for colorectal cancer (CRC) are associated with significant side effects and limitations, driving the search for novel therapeutic approaches. This study investigated the antiproliferative potential of D-limonene, a natural compound, on human colorectal adenocarcinoma (Caco-2) cells and analyzed its underlying mechanisms. Caco-2 cells were treated with D-limonene or doxorubicin (DOX) for 24 h. Cell viability was assessed using the MTT assay, with D-limonene and DOX showing IC50 values of 18.6 and 6.4 µM, respectively. In comparison to controls, D-limonene treatment dramatically enhanced the formation of reactive oxygen species (ROS) and decreased cellular antioxidant capacity, as seen by concentration-dependent lower glutathione (GSH) levels. The substance also increased the levels of pro-apoptotic proteins (caspase-3, Bax), tumor suppressor p53, lactate dehydrogenase (LDH), and inflammatory indicators [tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1β)]. Furthermore, in a concentration-dependent way, D-limonene therapy decreased the levels of matrix metalloproteinases (MMP2, MMP9), proliferation marker Ki67, and the anti-apoptotic protein Bcl-2. These results imply that the induction of oxidative stress, inflammation, and apoptotic pathways mediates D-limonene’s antiproliferative actions in colon cancer cells. Our findings show that D-limonene has therapeutic promise as a natural substitute for the treatment of colorectal cancer. Full article
(This article belongs to the Special Issue Natural Compounds: An Adjuvant Strategy in Cancer Management)
Show Figures

Graphical abstract

16 pages, 5364 KiB  
Article
Probiotic Administration Modulates Gut Microbiota and Suppresses Tumor Growth in Murine Models of Colorectal Cancer
by Anna Niechcial, Marlene Schwarzfischer, Paulina Wawrzyniak, Madita Determann, Doris Pöhlmann, Marcin Wawrzyniak, Emilie Gueguen, Maria R. Walker, Yasser Morsy, Kirstin Atrott, Marijn Wilmink, Luise Linzmeier, Marianne R. Spalinger, Sophie Holowacz, Anne Leblanc and Michael Scharl
Int. J. Mol. Sci. 2025, 26(9), 4404; https://doi.org/10.3390/ijms26094404 - 6 May 2025
Viewed by 1199
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide with limited treatment options for advanced disease stages. Growing evidence implicates the gut microbiota in CRC pathogenesis, prompting interest in probiotics as a potential therapeutic strategy. In this study, we evaluated the [...] Read more.
Colorectal cancer (CRC) is a leading cause of cancer-related mortality worldwide with limited treatment options for advanced disease stages. Growing evidence implicates the gut microbiota in CRC pathogenesis, prompting interest in probiotics as a potential therapeutic strategy. In this study, we evaluated the effects of two probiotic compositions, CI (a mix of lactobacilli and bifidobacteria) and CII (bifidobacteria alone), in two murine CRC models: the orthotopic MC-38 cecum injection model and the inflammation-driven azoxymethane/dextran sodium sulfate (AOM/DSS) model. CI showed significant anti-tumor effects in the orthotopic model, reducing tumor weight and volume, which was, however, not associated with robust immune activation, suggesting microbiota-driven mechanisms. In contrast, CII was more effective in the AOM/DSS model, reducing colonic inflammation and completely preventing tumor development. Our study demonstrates that probiotics might have great therapeutic potential via modulation of the gut microbiota, and they can exert anti-tumor effects in murine models of CRC with distinct compositions showing differential efficacy depending on the model. CI stabilized the gut microbiome and inhibited pro-tumorigenic taxa in the MC-38 cecum injection model, while CII exhibited anti-inflammatory properties in the AOM/DSS model, highlighting the potential of probiotics as context-specific interventions for CRC. These findings contribute to the growing body of evidence supporting microbiota-targeted strategies in oncology and their relevance for therapeutic applications. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Bioactive Nutrients Promoting Human Health)
Show Figures

Figure 1

30 pages, 6820 KiB  
Article
Propranolol and Capecitabine Synergy on Inducing Ferroptosis in Human Colorectal Cancer Cells: Potential Implications in Cancer Therapy
by Shiekhah Mohammad Alzahrani, Huda Abdulaziz Al Doghaither, Hind Ali Alkhatabi, Mohammad Abdullah Basabrain and Peter Natesan Pushparaj
Cancers 2025, 17(9), 1470; https://doi.org/10.3390/cancers17091470 - 27 Apr 2025
Cited by 1 | Viewed by 957
Abstract
Background/Objectives: Colorectal cancer (CRC) is a significant global health issue with rising incidence and mortality rates. In oncology, drug repurposing has emerged as a promising therapeutic strategy in conjunction with conventional treatments. This study aimed to evaluate the potential of repurposing propranolol (PRO), [...] Read more.
Background/Objectives: Colorectal cancer (CRC) is a significant global health issue with rising incidence and mortality rates. In oncology, drug repurposing has emerged as a promising therapeutic strategy in conjunction with conventional treatments. This study aimed to evaluate the potential of repurposing propranolol (PRO), a beta blocker, for the treatment of CRC cell lines (HCT-116 and HT-29), both as a monotherapy and in combination with capecitabine (CAP). Methods: Effects of mono- and combination therapies on viability, combination index, morphology, and cell death induction of CRC cells were assessed. Transcriptome analysis of HT-29 cells was performed using RNA sequencing. Metabolite profiling was conducted, and changes in biochemical parameters were evaluated using flow cytometry and biochemical analyses. Results: The combination index showed that HT-29 cells were the most responsive to the combined treatment, even with PIK3CA, B-RAF (V600E), and TP53 mutations. Moreover, ferroptosis was synergistically activated in the combined group of HT-29 in comparison to control. Furthermore, we observed an increase in OXPHOS metabolites, along with elevated intracellular and mitochondrial ROS, disruption of mitochondrial membrane potential, and greater levels of malondialdehyde (MDA) in the HT-29 combined group, which are the features of ferroptosis. Furthermore, ferroptosis induction was coupled with necroptosis, as indicated by RNA-sequencing data. Combination therapy inhibited cell migration and enhanced the immune response of HT-29 cells. Conclusions: These findings suggest that PRO is promising as a potential adjuvant therapy in combination with CAP for the treatment of CRC. Only HT-29 cells with the B-RAF (V600E) mutation showed promising findings in this study. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

Back to TopTop