Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,759)

Search Parameters:
Keywords = tumorigenic

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 5080 KB  
Article
Apigenin Induces Autophagy and Apoptosis in Chemoresistant Glioblastoma Cells and Inhibits Tumorigenicity Associated with Regulation of Immunomodulatory Proteins and Glial Cells Response
by Paulo Lucas Cerqueira Coelho, Cleonice Creusa dos Santos, Alessandra Bispo da Silva, Karina Costa da Silva, Monique Reis de Santana, Balbino Lino dos Santos, Giselle Pinto de Faria Lopes, Marie Pierre Junier, Hervé Chneiweiss, Vivaldo Moura-Neto, Maria de Fátima Dias Costa, Suzana Braga-de-Souza and Silvia Lima Costa
Cells 2025, 14(19), 1552; https://doi.org/10.3390/cells14191552 - 3 Oct 2025
Viewed by 309
Abstract
Background: Glioblastomas (GBMs) are the most aggressive and common neoplasms that affect glial cells, presenting rapid growth, invasion, and resistance to treatments. Studies have demonstrated the potentially inhibitory effect of flavonoids on glioblastoma cells’ stemness and viability. However, further research is needed to [...] Read more.
Background: Glioblastomas (GBMs) are the most aggressive and common neoplasms that affect glial cells, presenting rapid growth, invasion, and resistance to treatments. Studies have demonstrated the potentially inhibitory effect of flavonoids on glioblastoma cells’ stemness and viability. However, further research is needed to explore sensitivity and the mechanism of action in chemoresistant cells. Methods: In this study, we characterized the impact of apigenin treatment on the viability and differentiation of human GBM cells in vitro and its effects on tumorigenesis and regulation of the inflammatory response in vivo. Results: The flavonoid apigenin reduced the viability of U-251 cells, patient-derived cells TG-1 and OB-1 stem cells in a dose-dependent manner, associated with the induction of acidic vesicle organelles formation and apoptosis. Treatment with apigenin also inhibited migration and induced neural differentiation in the remaining viable cells, characterized by a decrease in the expression of the precursor marker nestin and an increase in the expression of astrocyte and neuron markers, GFAP and β-III tubulin, respectively. The xenotransplantation of apigenin-pretreated U251 cells into rat brains did not lead to tumor formation, unlike untreated cells. The surrounding area of transplanted untreated U251 cells exhibited reactive microglia and astrocytes, along with increased VEGF expression, which was absent in implant sites of apigenin-pretreated GBM cells. Moreover, in this implant area, we observed a significant decrease in the expression of mRNA for inflammatory factors IL-1β, TNF, and NOS2, and the downregulation of IL-10 and IL-4. Conclusions: These results demonstrate that apigenin inhibits the growth of tumoral cells, affecting the viability of tumor stem cells and impairing tumorigenicity, while altering the regulatory profile of immunomodulatory proteins. Therefore, this flavonoid can be considered for further studies to determine its use as an adjuvant to the treatment of human GBMs. Full article
(This article belongs to the Special Issue The Pivotal Role of Tumor Stem Cells in Glioblastoma: Second Edition)
Show Figures

Figure 1

18 pages, 8074 KB  
Article
Auranofin Ameliorates Gouty Inflammation by Suppressing NLRP3 Activation and Neutrophil Migration via the IL-33/ST2–CXCL1 Axis
by Hyeyeon Yoo, Ahyoung Choi, Minjun Kim, Yongseok Gye, Hyeonju Jo, Seung-Ki Kwok, Youngjae Park and Jennifer Jooha Lee
Cells 2025, 14(19), 1541; https://doi.org/10.3390/cells14191541 - 2 Oct 2025
Viewed by 331
Abstract
Gout is a form of sterile inflammatory arthritis in which monosodium urate (MSU) crystals deposit and provoke a neutrophil-predominant response, primarily driven by activation of the NACHT, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome. Here, we show that auranofin, a Food [...] Read more.
Gout is a form of sterile inflammatory arthritis in which monosodium urate (MSU) crystals deposit and provoke a neutrophil-predominant response, primarily driven by activation of the NACHT, leucine-rich repeat, and pyrin domain-containing protein 3 (NLRP3) inflammasome. Here, we show that auranofin, a Food and Drug Administration (FDA)-approved anti-rheumatic agent, exerts anti-inflammatory effects in both in vitro and in vivo models of gout. Auranofin inhibited NLRP3 inflammasome activation in human THP-1 cells and murine macrophages, leading to reduced cleavage of caspase-1, interleukin-1β (IL-1β), and interleukin-18 (IL-18). In MSU crystal-induced mouse models, auranofin treatment reduced paw swelling, serum cytokine levels, and tissue inflammation. Notably, auranofin suppressed neutrophil migration and decreased expression of C-X-C motif chemokine ligand 1 (CXCL1) in inflamed foot tissue and air-pouch exudates. Mechanistically, auranofin disrupted the interleukin-33 (IL-33)/suppression of tumorigenicity 2 (ST2) axis, a key signaling pathway promoting neutrophil recruitment. Overexpression of IL-33 abolished the anti-inflammatory effects of auranofin, highlighting the central role of IL-33 in gout pathogenesis. Together, our findings suggest that auranofin alleviates MSU-induced inflammation by concurrently inhibiting NLRP3 inflammasome activation and IL-33-mediated neutrophil recruitment, supporting its potential as a dual-action therapeutic candidate for gout. Full article
(This article belongs to the Section Cellular Immunology)
Show Figures

Graphical abstract

14 pages, 2614 KB  
Article
Brassica Extracts Prevent Benzo(a)pyrene-Induced Transformation by Modulating Reactive Oxygen Species and Autophagy
by José Benito Montes-Alvarado, Paula Garcia-Ibañez, Diego A. Moreno, Fabiola Lilí Sarmiento-Salinas, Xiadani Edén Susano-Hernández, Karen Andrea Larrauri-Rodríguez, Francisco Jesús García-Hernández, Lorena Milflores-Flores, Fabiola Domínguez and Paola Maycotte
Int. J. Mol. Sci. 2025, 26(19), 9519; https://doi.org/10.3390/ijms26199519 - 29 Sep 2025
Viewed by 155
Abstract
Plants from the Brassicaceae family are characterized by their high content of glucosinolates (GSLs), whose hydrolysis products, isothiocyanates (ITC) or indole compounds, have been found to have anti-inflammatory, antioxidant and metabolic regulatory functions. In this work, we used a model of transformation using [...] Read more.
Plants from the Brassicaceae family are characterized by their high content of glucosinolates (GSLs), whose hydrolysis products, isothiocyanates (ITC) or indole compounds, have been found to have anti-inflammatory, antioxidant and metabolic regulatory functions. In this work, we used a model of transformation using the MCF10A cell line, a non-tumorigenic breast fibrocystic disease cell line, treated with benzo(a)pyrene (B(a)P), a potent carcinogen known to induce the production of reactive oxygen species (ROS) and DNA damage. Broccoli sprout (BSE) or red cabbage aqueous (RCA) extracts were rich in ITC and indole compounds. Their use decreased B(a)P induced cellular proliferation and ROS production. in addition, RCA extract induced autophagy in MCF10A cells. Our results indicate a potential use of BSE or RCA for the prevention of carcinogen-induced transformation and of RCA as a method for autophagy, a tumor suppressor pathway, induction. Full article
(This article belongs to the Special Issue Bioactive Compounds in the Prevention of Chronic Diseases)
Show Figures

Figure 1

27 pages, 3718 KB  
Review
The Impact of Helminths on Colorectal Cancer: From Infections to the Isolation of Biotherapeutics
by Cuauhtémoc Ángel Sánchez-Barrera, Karen V. Fernandez-Muñoz, Mónica G. Mendoza-Rodríguez, María T. Ortiz-Melo, Jazmín A. Carrillo-Pérez, Miriam Rodríguez-Sosa and Luis I. Terrazas
Pathogens 2025, 14(9), 949; https://doi.org/10.3390/pathogens14090949 - 20 Sep 2025
Viewed by 484
Abstract
Worldwide, colorectal cancer (CRC) is the third-most common cancer and the second-leading cause of cancer-related deaths. The inflammatory response initiated by pathogens, environmental and dietary factors, and inflammatory bowel diseases can promote the formation of colorectal tumors. The hygiene hypothesis proposes an inverse [...] Read more.
Worldwide, colorectal cancer (CRC) is the third-most common cancer and the second-leading cause of cancer-related deaths. The inflammatory response initiated by pathogens, environmental and dietary factors, and inflammatory bowel diseases can promote the formation of colorectal tumors. The hygiene hypothesis proposes an inverse link between inflammatory diseases and early childhood exposure to pathogens, with a significant negative correlation between chronic inflammatory diseases and helminth infections. On the other hand, it is also known that several pathogens may influence or even cause the development of cancer, including helminth infections. How do helminth infections influence CRC outcomes? The existing literature presents two different perspectives. Experimental studies in CRC models suggest that helminths may accelerate disease progression and lead to worse outcomes (such as Schistosoma and Trichuris sp.), while others indicate that helminths could help reduce tumor burden (such as Taenia sp.). This review focuses on helminths’ pro- and anti-tumorigenic effects and their derivatives, specifically in CRC. We provide a comprehensive understanding of how helminths impact the macroscopic, histopathological, immunological, and molecular aspects of CRC. Full article
(This article belongs to the Special Issue Immunity and Immunoregulation in Helminth Infections)
Show Figures

Graphical abstract

17 pages, 935 KB  
Review
Utilization of AhR and GPR35 Receptor Ligands as Superfoods in Cancer Prevention for Individuals with IBD
by Olga Poźniak, Robert Sitarz, Monika Zofia Sitarz, Dorota Kowalczuk, Emilia Słoń and Ewa Dudzińska
Int. J. Mol. Sci. 2025, 26(18), 9160; https://doi.org/10.3390/ijms26189160 - 19 Sep 2025
Viewed by 439
Abstract
Carcinogenesis is a complex process characterized by the uncontrolled proliferation of abnormal cells, influenced by environmental, genetic, and epigenetic factors. Chronic inflammation is undoubtedly one of the key contributors to carcinogenesis. Inflammatory bowel disease (IBD) is associated with an increased risk of colorectal [...] Read more.
Carcinogenesis is a complex process characterized by the uncontrolled proliferation of abnormal cells, influenced by environmental, genetic, and epigenetic factors. Chronic inflammation is undoubtedly one of the key contributors to carcinogenesis. Inflammatory bowel disease (IBD) is associated with an increased risk of colorectal cancer (CRC) due to persistent inflammation resulting from continuous immune system activation and excessive immune cell recruitment. IBD is also linked to certain nutritional deficiencies, primarily due to dietary modifications necessitated by the disease’s pathophysiology. Consequently, individualized nutritional supplementation appears to be a rational approach to addressing these deficiencies. The use of functional foods, including anti-inflammatory nutraceuticals, in individuals with IBD may play a crucial role in modulating cellular pathways that inhibit the release of inflammatory mediators. Thus, the regulation of the aryl hydrocarbon receptor (AhR) and G protein-coupled receptor 35 (GPR35) through dietary ligands appears to be of significant importance not only in the treatment of IBD and maintenance of remission but also in the prevention of tumorigenic transformation, particularly in genetically predisposed individuals. This narrative review was conducted using PubMed, Scopus, and Web of Science databases. The search covered literature published between January 2000 and June 2024. Keywords included ‘inflammatory bowel disease’, ‘colorectal cancer’, ‘AhR’, ‘aryl hydrocarbon receptor’, ‘GPR35’, ‘cytochrome P450’, ‘nutraceuticals’, ‘probiotics’, and ‘superfoods’. Only English-language articles were included. The selection focused on studies investigating mechanistic pathways and the role of dietary ligands in AhR and GPR35 activation in IBD and CRC. The SANRA guidelines for narrative reviews were followed to ensure transparency and minimize bias. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Figure 1

16 pages, 1387 KB  
Review
Roles of Neutrophils in Autoimmune Diseases and Cancers
by Anjali Bhargav, Vinay Kumar and Neeraj Kumar Rai
Int. J. Mol. Sci. 2025, 26(18), 9040; https://doi.org/10.3390/ijms26189040 - 17 Sep 2025
Viewed by 541
Abstract
Neutrophils, a first-line defender, has a multifaceted presence in chronic inflammation, autoimmune pathology, and tumor progression. The microenvironmental cues facilitate functional plasticity and phenotypic heterogeneity to neutrophils that enable both their protective and pathogenic roles. Autoimmune diseases including systemic lupus erythematosus (SLE), rheumatoid [...] Read more.
Neutrophils, a first-line defender, has a multifaceted presence in chronic inflammation, autoimmune pathology, and tumor progression. The microenvironmental cues facilitate functional plasticity and phenotypic heterogeneity to neutrophils that enable both their protective and pathogenic roles. Autoimmune diseases including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), and juvenile idiopathic arthritis (JIA) display the presence of dysregulated subsets of neutrophil, such as low-density granulocytes (LDGs) that promote proinflammation and contribute to tissue damage via NETosis and type I interferon-mediated signaling. In cancer, particularly tumors, they exhibit tumor-associated neutrophils (TANs) which may polarize either towards anti-tumorigenic ‘N1’ or pro-tumorigenic ‘N2’ phenotypes based on available modulators such as TGF-β and leucine-driven epigenetic modifications. The development in neutrophil biology has introduced several novel therapeutic strategies that allow NET targeting, inhibition of chemokine receptors like CXCR2, and exploration of neutrophil-derived biomarkers for diagnosis and disease monitoring. Such findings encourage the importance of neutrophils as both effectors and therapeutic targets in inflammatory and neoplastic conditions. Full article
Show Figures

Figure 1

25 pages, 410 KB  
Review
Beneficial Handling of Molecular Chaperones (Chaperonotherapy) in Glioblastoma and Neuroblastoma: Novel Therapeutic Targets or Potential Agents?
by Maria Antonella Augello, Nima Shadan, Giuseppa D’Amico, Rosario Barone, Celeste Caruso Bavisotto, Federica Scalia and Alessandra Maria Vitale
Cells 2025, 14(18), 1447; https://doi.org/10.3390/cells14181447 - 16 Sep 2025
Viewed by 684
Abstract
Molecular chaperones, especially Heat Shock Proteins (HSPs), play complex, context-dependent roles in cancer, particularly in nervous system (NS) tumors like glioblastoma (GBM) and neuroblastoma (NB). They are often upregulated, promoting tumor growth, poor prognosis, and resistance to therapy and immune responses. This supports [...] Read more.
Molecular chaperones, especially Heat Shock Proteins (HSPs), play complex, context-dependent roles in cancer, particularly in nervous system (NS) tumors like glioblastoma (GBM) and neuroblastoma (NB). They are often upregulated, promoting tumor growth, poor prognosis, and resistance to therapy and immune responses. This supports the potential of negative chaperonotherapy, aimed at inhibiting them. However, some studies suggest chaperones can also act as tumor suppressors in certain cancers, indicating that positive chaperonotherapy—enhancing or restoring their function—may be beneficial. For NS tumors, this latter area is still understudied. With emphasis on GBM and NB, in this review we address the potential of molecular chaperones, particularly HSPs, as therapeutic targets or agents. We discuss strategies to inhibit pro-tumorigenic chaperones as well as the underexplored potential of chaperone induction and immunomodulation. Ultimately, we examine the emerging use of pharmacological and chemical chaperones to improve treatment outcomes in these NS tumors. These strategies, whether applied alone or in combination, may offer significant benefits for GBM and NB, which are presently among the most aggressive and challenging tumors to manage. Full article
Show Figures

Graphical abstract

38 pages, 4420 KB  
Article
Uncovering the Tumorigenic Blueprint of PFOS and PFOA Through Multi-Organ Transcriptomic Analysis of Biomarkers, Mechanisms, and Therapeutic Targets
by Krisha Mathur, Aleezah Khaliq, Stephanie Park, Nathan Chu, Vaishnavi M. Burra, Norah Kanukolanu, Ellen Costello and Sivanesan Dakshanamurthy
Curr. Issues Mol. Biol. 2025, 47(9), 763; https://doi.org/10.3390/cimb47090763 - 15 Sep 2025
Viewed by 1022
Abstract
Per- and polyfluoroalkyl substances (PFASs), called forever chemicals, persist in the environment and bioaccumulate, posing significant health risks. While epidemiological studies have linked exposure to specific PFAS types, perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS), to an increased incidence of various cancers, [...] Read more.
Per- and polyfluoroalkyl substances (PFASs), called forever chemicals, persist in the environment and bioaccumulate, posing significant health risks. While epidemiological studies have linked exposure to specific PFAS types, perfluorooctanoic acid (PFOA) and perfluorooctane sulfonic acid (PFOS), to an increased incidence of various cancers, specific tumorigenesis mechanisms are unknown. Here, we investigated the potential molecular markers and signatures of perfluorooctane sulfonic acid (PFOS) and perfluorooctanoic acid (PFOA) tumorigenesis. We performed a comprehensive transcriptomic analysis across multiple species and tissue types (N = 529) using PFOS and PFOA-exposed RNA-Seq samples. Conserved signatures demonstrate significant disruptions in seven key carcinogenic characteristics including metabolic reprogramming, epigenetic modifications, immune suppression, oxidative stress, and genomic instability. Tumorigenic markers such as SERPINE1, FN1, PLIN2, ALDOA, TRIB3, and TSC22D3 and their associated pathways may act independently or synergistically to promote a pro-tumorigenic environment. Additionally, PPARα, LARP1, ACOX1, MYC, and MYCN were identified as key upstream regulators supporting disruptions in lipid metabolism, oxidative stress, and uncontrolled cell proliferation. In liver samples, low concentrations of PFOS and PFOA were sufficient to exhibit tumorigenic signatures associated with tumorigenesis initiation and development. Inferred mechanisms of ccRCC initiation and development were linked to lipid metabolism dysregulation and immunosuppressive signaling. In prostate and testicular xenograft tumor models, carcinogenic mechanisms for tumor progression and promotion were hypothesized. Receptor-mediated signaling and protein synthesis was disrupted in prostate cancer and epigenetic alterations and ECM remodeling observed in testicular cancer. We also explored potential therapeutic rescue strategies, including chemopreventive agents for early intervention. All our findings provide hypotheses for PFOS/PFOA-induced tumorigenesis; however, experimental studies are required to establish translational relevance. All the R codes developed in this study are publicly available. Full article
(This article belongs to the Special Issue Genomic Analysis of Common Disease, 2nd Edition)
Show Figures

Graphical abstract

27 pages, 5855 KB  
Article
Derivation of Genetically Defined Murine Hepatoblastoma Cell Lines with Angiogenic Potential
by Keyao Chen, Ahmet Toksoz, Colin Henchy, Jessica Knapp, Jie Lu, Sarangarajan Ranganathan, Huabo Wang and Edward V. Prochownik
Cancers 2025, 17(18), 3002; https://doi.org/10.3390/cancers17183002 - 14 Sep 2025
Viewed by 489
Abstract
Background/Objectives: Hepatoblastoma (HB), the most common pediatric liver cancer, often bears mutations in and/or otherwise deregulates the oncogenic transcription factors β-catenin (B), YAP (Y) and NRF2 (N). HB research is hampered by a paucity of established cell lines, particularly those possessing these [...] Read more.
Background/Objectives: Hepatoblastoma (HB), the most common pediatric liver cancer, often bears mutations in and/or otherwise deregulates the oncogenic transcription factors β-catenin (B), YAP (Y) and NRF2 (N). HB research is hampered by a paucity of established cell lines, particularly those possessing these molecular drivers. All combinations of B, Y and N (BY, BN, YN and BYN) are tumorigenic when overexpressed in murine livers, but it has not been possible to establish cell lines from primary tumors. Recently, we found that concurrent, in vivo Crispr-mediated targeting of the Cdkn2a tumor suppressor locus allows for immortalized cell lines to be efficiently generated. Methods: We derived and characterized five immortalized cell lines from Cdkn2a-targeted BN and YN HBs. Results: Four of the above five cell lines retained their ability to grow as subcutaneous or “pseudo-metastatic” pulmonary tumors in the immunocompetent mice from which they originated. Most notably, when maintained under hypoxic conditions for as little as 2 days, BN cells transiently upregulated the expression of numerous endothelial cell (EC)-specific genes and acquired EC-like properties that benefited tumor growth. These lines and those from previously derived BY and BYN HBs also possessed similar sensitivities to four commonly employed chemotherapeutic drugs. Conclusions: The above-described approach is currently the only means to generate HB cell lines with pre-selected and clinically relevant oncogenic drivers. Its generic nature should also allow bespoke HB cell lines with other oncogenic drivers to be readily produced. A collection of such cell lines will be useful for studying tumor cell-to-EC trans-differentiation, interactions with the immune environment and drug sensitivities. Full article
(This article belongs to the Section Molecular Cancer Biology)
Show Figures

Graphical abstract

25 pages, 8714 KB  
Article
Cell Line-Dependent Internalization, Persistence, and Immunomodulatory Effects of Staphylococcus aureus in Triple-Negative Breast Cancer
by Sima Kianpour Rad, Runhao Li, Kenny K. L. Yeo, Clare Cooksley, Gohar Shaghayegh, Sarah Vreugde, Fangmeinuo Wu, Yoko Tomita, Timothy J. Price, Wendy V. Ingman, Amanda R. Townsend and Eric Smith
Cancers 2025, 17(18), 2947; https://doi.org/10.3390/cancers17182947 - 9 Sep 2025
Viewed by 1349
Abstract
Background: Triple-negative breast cancer (TNBC) is an aggressive subtype with limited therapeutic options and inconsistent response to immune checkpoint inhibitors (ICIs). Emerging evidence indicates that tumor-associated bacteria may shape immune signaling and alter immunotherapy outcomes. Here, we investigated whether Staphylococcus aureus invades TNBC [...] Read more.
Background: Triple-negative breast cancer (TNBC) is an aggressive subtype with limited therapeutic options and inconsistent response to immune checkpoint inhibitors (ICIs). Emerging evidence indicates that tumor-associated bacteria may shape immune signaling and alter immunotherapy outcomes. Here, we investigated whether Staphylococcus aureus invades TNBC cells, persists intracellularly, and modulates PD-L1 expression. Methods: Using eFluor450-labeled S. aureus for flow cytometry, gentamicin protection assays, CFU quantification, and transmission electron microscopy, we assessed bacterial uptake and persistence in six TNBC cell lines and a non-tumorigenic control. PD-L1, TLR2, and STAT1 activation were evaluated after infection or TLR2 ligand treatment ± IFN-γ. Results: At multiplicity of infection (MOI) of 10, S. aureus internalized into 67% of MDA-MB-468 and 54% of MDA-MB-231, with intermediate uptake in Hs578T (27%) and BT-549 (24%) and only 0.5–9% in low-uptake lines (MDA-MB-453, CAL-51, MCF-12A). High-uptake lines exhibited marked cytotoxicity and reduced proliferation, with MDA-MB-468 showing an 82% drop in viability at 2 h and a 74% decrease after 5 d, whereas low-uptake lines showed minimal impact. Persistence lasted >7 d in MDA-MB-231 but only 3–5 days in others. IFN-γ plus S. aureus significantly amplified PD-L1, with up to a 2.9-fold increase in MDA-MB-468 and 1.5-fold in MDA-MB-231, but no effect in low-uptake lines. TLR2 agonists modestly increased PD-L1 in high-TLR2-expressing lines and synergized with IFN-γ. These effects were accompanied by STAT1 phosphorylation, supporting a TLR2/STAT1 axis linking bacterial sensing to immune checkpoint regulation. Conclusions: Together, these findings identify S. aureus as a modulator of immune signaling in TNBC and highlight the potential for microbial factors to influence ICI responsiveness. Targeting tumor–bacteria interactions may represent a novel strategy to enhance immunotherapy efficacy in breast cancer. Full article
(This article belongs to the Special Issue New Insights into Microbial-Based Cancer Therapy)
Show Figures

Graphical abstract

19 pages, 6972 KB  
Article
Development and Characterization of a Novel Lineage of Renal Progenitor Cells for Potential Use in Feline Chronic Kidney Disease: A Preliminary Study
by Lara Carolina Mario, Juliana de Paula Nhanharelli, Jéssica Borghesi, Rafaela Rodrigues Ribeiro, Hianka Jasmyne Costa de Carvalho, Thamires Santos da Silva, Mariano del Sol, Rodrigo da Silva Nunes Barreto, Sandra Maria Barbalho and Maria Angelica Miglino
Cells 2025, 14(17), 1395; https://doi.org/10.3390/cells14171395 - 6 Sep 2025
Viewed by 694
Abstract
Chronic kidney disease (CKD) is a common and serious condition in felines. Accordingly, several cell therapies have been studied over the past decades for effective treatments. This study aimed to develop a new lineage of renal progenitor cells for use in cats with [...] Read more.
Chronic kidney disease (CKD) is a common and serious condition in felines. Accordingly, several cell therapies have been studied over the past decades for effective treatments. This study aimed to develop a new lineage of renal progenitor cells for use in cats with CKD. Metanephric and mesonephric progenitor cells were obtained from mesonephros and metanephros tissues of feline conceptuses at four distinct gestational stages. The cultured cells were characterized by their morphology, tumorigenic potential, immunophenotype determined by flow cytometry, and differentiation potential. We then conducted a pilot study in CKD-affected cats, comparing intraperitoneal injections of cultured metanephric progenitor cells (n = 4) to a placebo solution (n = 3). All four cell types exhibited adhesion and colony formation, but showed no tumorigenic potential. Cells tested positive for renal progenitor markers (CD117, Nephron, and WT1), confirming their identity. Treated cats showed no statistically significant differences (p ≤ 0.05) in any of the data analyzed. However, caregivers reported a voluntary increase in appetite after cell administration. Veterinarians confirmed this information during double-blind evaluations conducted after treatment. Although this data are qualitative, no clinical deterioration was observed in cats. Our results suggest that this new lineage of renal progenitor cells did not induce immediate adverse effects, thus supporting its potential for use in cell-based therapies. However, further studies are needed to evaluate its efficacy in treating renal diseases. Full article
(This article belongs to the Special Issue New Advances in Tissue Engineering and Regeneration)
Show Figures

Figure 1

21 pages, 2159 KB  
Review
The Interleukin-8-CXCR1/2 Axis as a Therapeutic Target in Peritoneal Carcinomatosis
by Christopher Sherry, Neda Dadgar, Zuqiang Liu, Yong Fan, Kunhong Xiao, Ali H. Zaidi, Vera S. Donnenberg, Albert D. Donnenberg, David L. Bartlett and Patrick L. Wagner
Curr. Oncol. 2025, 32(9), 496; https://doi.org/10.3390/curroncol32090496 - 5 Sep 2025
Viewed by 767
Abstract
Peritoneal carcinomatosis (PC) is a late-stage manifestation of abdominopelvic malignancies with poor prognosis and limited treatment options. Unique biochemical mechanisms within the peritoneal cavity play a key role in disease progression and resistance to therapy. Despite current therapies like systemic chemotherapy and cytoreductive [...] Read more.
Peritoneal carcinomatosis (PC) is a late-stage manifestation of abdominopelvic malignancies with poor prognosis and limited treatment options. Unique biochemical mechanisms within the peritoneal cavity play a key role in disease progression and resistance to therapy. Despite current therapies like systemic chemotherapy and cytoreductive surgery, patients frequently develop severe complications, including bowel obstruction, nutritional decline, and ascites, driving the need to address the pro-tumorigenic niche in the peritoneal cavity. The immune microenvironment in PC is marked by elevated proinflammatory mediators, such as IL-6 and IL-8, which skew the response toward innate rather than adaptive immune responses. IL-8 signaling, through its receptors CXCR1 and CXCR2, promotes neutrophil recruitment, chronic inflammation, angiogenesis, epithelial–mesenchymal transition, and immune evasion, making the IL-8/CXCR1/CXCR2 axis a potential therapeutic target in PC. Pre-clinical models provide evidence that IL-8 or CXCR1/CXCR2 blockade may be a valuable therapeutic strategy. IL-8 targeting agents such as monoclonal antibodies (BMS-986253) and small-molecule inhibitors (SX-682, AZD5069, navarixin) have shown efficacy in mitigating tumor growth and improving the efficacy of immune checkpoint inhibitors. Phase I and II trials have demonstrated encouraging safety profiles and preliminary efficacy when treating multiple abdominopelvic malignancies. In this review, we discuss the influence of the IL-8/CXCR1/CXCR2 axis within the peritoneal immune environment in PC and highlight recent work using IL-8 or CXCR1/CXCR2 blockade as a therapeutic strategy for PC. Continued research into the peritoneal immune microenvironment and the development of targeted therapies are essential for improving the management and prognosis of PC, potentially enhancing antitumor immunity and patient outcomes. Full article
(This article belongs to the Section Gastrointestinal Oncology)
Show Figures

Graphical abstract

17 pages, 3777 KB  
Article
Heparanase-Neutralizing Monoclonal Antibody (mAb A54) Attenuates Tumor Growth and Metastasis
by Uri Barash, Malik Farhoud, Maali Odeh, Eliezer Huberman, Liang Wu and Israel Vlodavsky
Cells 2025, 14(17), 1379; https://doi.org/10.3390/cells14171379 - 4 Sep 2025
Viewed by 787
Abstract
Heparanase is the only human enzyme responsible for heparan sulfate (HS) breakdown, an activity that remodels the extracellular matrix (ECM) and strongly drives cancer metastasis and angiogenesis. Compelling evidence implies that heparanase promotes essentially all aspects of the tumorigenic process, namely, tumor initiation, [...] Read more.
Heparanase is the only human enzyme responsible for heparan sulfate (HS) breakdown, an activity that remodels the extracellular matrix (ECM) and strongly drives cancer metastasis and angiogenesis. Compelling evidence implies that heparanase promotes essentially all aspects of the tumorigenic process, namely, tumor initiation, vascularization, growth, metastasis, and chemoresistance. A key mechanism by which heparanase accelerates cancer progression is by enabling the release and bioavailability of HS-bound growth factors, chemokines, and cytokines, residing in the tumor microenvironment and supporting tumor growth and metastasis. The currently available heparanase inhibitors are mostly HS/heparin-like compounds that lack specificity and exert multiple off-target side effects. To date, only four such compounds have progressed to clinical trials, and none have been approved for clinical use. We have generated and characterized an anti-heparanase monoclonal antibody (A54 mAb) that specifically inhibits heparanase enzymatic activity (ECM degradation assay) and cellular uptake. Importantly, A54 mAb attenuates xenograft tumor growth and metastasis (myeloma, glioma, pancreatic, and breast carcinomas) primarily when administered (syngeneic or immunocompromised mice) in combination with conventional anti-cancer drugs. Co-crystallization of the A54 Fab fragment and the heparanase enzyme revealed that the interaction between the two proteins takes place adjacent to the enzyme HS/heparin binding domain II (HBDII; Pro271-Ala276), likely hindering heparanase from interacting with HS substrates via steric occlusion of the active site cleft. Collectively, we have generated and characterized a novel mAb that specifically neutralizes heparanase enzymatic activity and attenuates its pro-tumorigenic effects in preclinical models, paving the way for its clinical examination against cancer, inflammation, and other diseases. Full article
Show Figures

Figure 1

22 pages, 1081 KB  
Review
Pseudouridine Synthase 7 in Cancer: Functions, Mechanisms, and Therapeutic Potential
by Qiwei Yang, Thomas G. Boyer and Ayman Al-Hendy
Cells 2025, 14(17), 1380; https://doi.org/10.3390/cells14171380 - 4 Sep 2025
Viewed by 998
Abstract
Pseudouridylation, the most abundant RNA modification, plays a critical role in modulating RNA structure, stability, and function. Among the family of pseudouridine synthases, Pseudouridine Synthase 7 (PUS7) has recently gained attention for its emerging roles in human health and disease. Originally characterized for [...] Read more.
Pseudouridylation, the most abundant RNA modification, plays a critical role in modulating RNA structure, stability, and function. Among the family of pseudouridine synthases, Pseudouridine Synthase 7 (PUS7) has recently gained attention for its emerging roles in human health and disease. Originally characterized for its function in modifying tRNA and small non-coding RNAs, PUS7 is now recognized as a dynamic regulator of mRNA pseudouridylation, influencing gene expression at the post-transcriptional level. Aberrant expressions or activity of PUS7 have been linked to a variety of pathological conditions, including cancers such as colon cancer, glioblastoma, pancreatic cancer, and neuroblastoma, as well as potential roles in neurodevelopmental disorders and immune regulation. Through mechanisms involving translational reprogramming, stress adaptation, and epitranscriptomic remodeling, PUS7 contributes to disease progression and cellular plasticity. This review summarizes the current understanding of PUS7 biology, its functional relevance in the contexts of cancer progression, and the growing interest in targeting RNA-modifying enzymes for therapeutic intervention. Uncovering the full spectrum of PUS7-mediated pseudouridylation and its downstream effects holds promise for advancing our understanding of RNA-based regulation in human diseases, including gynecological disorders. Full article
Show Figures

Figure 1

32 pages, 962 KB  
Review
Digital Twin-Based Multiscale Models for Biomarker Discovery in Kinase and Phosphatase Tumorigenic Processes
by Sara Sadat Aghamiri and Rada Amin
Kinases Phosphatases 2025, 3(3), 18; https://doi.org/10.3390/kinasesphosphatases3030018 - 31 Aug 2025
Viewed by 631
Abstract
Digital twin is a mathematical model that virtually represents a physical object or process and predicts its behavior at future time points. These simulation models enable a deeper understanding of tumorigenic processes and improve biomarker discovery in cancer research. Tumor microenvironment is marked [...] Read more.
Digital twin is a mathematical model that virtually represents a physical object or process and predicts its behavior at future time points. These simulation models enable a deeper understanding of tumorigenic processes and improve biomarker discovery in cancer research. Tumor microenvironment is marked by dysregulated signaling pathways, where kinases and phosphatases serve as critical regulators and promising sources for biomarker discovery. These enzymes operate within multiscale and context-dependent processes where spatial and temporal coordination determine cellular outcomes. Digital Twin technology provides a platform for multimodal and multiscale modeling of kinase and phosphatase processes at the patient-specific level. These models have the potential to transform biomarker validation processes, enhance the prediction of therapeutic responses, and support precision decision-making. In this review, we present the major alterations affecting kinases and phosphatase functions within the tumor microenvironment and their clinical relevance as biomarkers, and we address how digital twins in oncology can augment and refine each stage of the biomarker discovery pipeline. Introducing this emerging technology for cancer biomarker discovery will assist in accelerating its adoption and translation into precision diagnostics and targeted therapies. Full article
Show Figures

Figure 1

Back to TopTop