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
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

Search Results (2,249)

Search Parameters:
Keywords = anti-cancer immune response

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
27 pages, 1567 KiB  
Review
NR4A1 Acts as a Nutrient Sensor That Inhibits the Effects of Aging
by Stephen Safe
Nutrients 2025, 17(16), 2709; https://doi.org/10.3390/nu17162709 - 21 Aug 2025
Viewed by 28
Abstract
Orphan nuclear receptor 4A1 (NR4A1) is a member of the NR4A subfamily that was initially discovered as an intermediate early gene expressed in response to stressors, including inflammatory agents. This review addresses the hypothesis that NR4A1 is a key nutrient sensor that contributes [...] Read more.
Orphan nuclear receptor 4A1 (NR4A1) is a member of the NR4A subfamily that was initially discovered as an intermediate early gene expressed in response to stressors, including inflammatory agents. This review addresses the hypothesis that NR4A1 is a key nutrient sensor that contributes to the anti-aging and health-protective effects of receptor ligands, dietary phenolics, and other diet-derived compounds. There is evidence in animal models including humans that NR4A1 serves as an important gene that decreases the rate of aging and its associated diseases. For example, in humans and mice, NR4A1 expression decreases with age and loss of NR4A1 enhances disease susceptibility, and survival curves show that NR4A1-deficient mice live 4 months less than wild-type animals. An extensive comparison of inflammatory diseases, immune dysfunction, and fibrosis in multiple tissues shows that in NR4A1−/− mice and rats these diseases and injuries are enhanced compared to wild-type NR4A1−/− animals. There is evidence showing that structurally diverse NR4A1 ligands reverse the induced adverse effects in NR4A1 wild-type mice. This raises an important question regarding the mechanisms of NR4A1-dependent inhibition of the aging process and the potential for this receptor as a nutrient sensor. It has been well established that polyphenolics, including flavonoids, resveratrol, and other compounds in the diet, are health-protective and decrease the aging process. Recent studies show that resveratrol and flavonoids such as quercetin and kaempferol bind NR4A1 and exhibit protective NR4A1-dependent inhibition of endometriosis and cancer. These limited studies support a role for NR4A1 as a potential dietary sensor of nutrients that are known to be health-protective and a potential nutrient target for improving health. Full article
(This article belongs to the Section Geriatric Nutrition)
Show Figures

Graphical abstract

31 pages, 2856 KiB  
Review
Therapeutic Potential of Propolis in Preclinical Models of Cancer and Infectious Diseases: A Review
by Michał K. Pierański, Damian Kaniowski and Piotr Szweda
Int. J. Mol. Sci. 2025, 26(16), 8041; https://doi.org/10.3390/ijms26168041 - 20 Aug 2025
Viewed by 193
Abstract
Propolis is a complex, resinous substance originating from plant exudates and processed by bees, e.g., Apis mellifera L. Propolis is rich in flavonoids, phenolic acids, and terpenoids. It exhibits broad biological activities, including antimicrobial, anti-inflammatory, immunomodulatory, and anticancer effects. This review summarizes recent [...] Read more.
Propolis is a complex, resinous substance originating from plant exudates and processed by bees, e.g., Apis mellifera L. Propolis is rich in flavonoids, phenolic acids, and terpenoids. It exhibits broad biological activities, including antimicrobial, anti-inflammatory, immunomodulatory, and anticancer effects. This review summarizes recent findings on the therapeutic potential of propolis in preclinical models of cancer and infectious diseases, with a focus on its molecular mechanisms of action. Experimental data indicate that propolis and its active constituents can induce apoptosis, inhibit proliferation, angiogenesis, and metastasis of cancer cells, and modulate immune responses and microbial virulence. Despite promising in vitro results, in vivo studies remain limited, and their results are often inconsistent. The variability in chemical composition due to geographical and botanical factors, as well as the lack of standardized extracts, further impedes translational research. We highlight key molecular pathways affected by propolis and propose directions for future studies, including improved standardization and more rigorous in vivo results description. These efforts are essential to validate propolis as a potential booster or alternative therapeutic strategy in oncology and infectious diseases treatment. Full article
(This article belongs to the Special Issue Role of Natural Products in Health and Diseases)
Show Figures

Figure 1

31 pages, 2786 KiB  
Review
Mechanisms and Therapeutic Advances of PXR in Metabolic Diseases and Cancer
by Yuanbo Bi, Sifan Liu, Lei Wang, Daiyin Peng, Weidong Chen, Yue Zhang and Yanyan Wang
Int. J. Mol. Sci. 2025, 26(16), 8029; https://doi.org/10.3390/ijms26168029 - 20 Aug 2025
Viewed by 114
Abstract
The pregnane X receptor (PXR), a ligand-activated nuclear receptor, plays a central role in regulating the metabolism of both endogenous substances and xenobiotics. In recent years, increasing evidence has highlighted its involvement in chronic diseases, particularly metabolic disorders and cancer. PXR modulates drug-metabolizing [...] Read more.
The pregnane X receptor (PXR), a ligand-activated nuclear receptor, plays a central role in regulating the metabolism of both endogenous substances and xenobiotics. In recent years, increasing evidence has highlighted its involvement in chronic diseases, particularly metabolic disorders and cancer. PXR modulates drug-metabolizing enzymes, transporters, inflammatory factors, lipid metabolism, and immune-related pathways, contributing to the maintenance of hepatic–intestinal barrier homeostasis, energy metabolism, and inflammatory responses. Specifically, in type 2 diabetes mellitus (T2DM), PXR influences disease progression by regulating glucose metabolism and insulin sensitivity. In obesity, it affects adipogenesis and inflammatory processes. In atherosclerosis (AS), PXR exerts protective effects through cholesterol metabolism and anti-inflammatory actions. In metabolic dysfunction-associated steatotic liver disease (MASLD), it is closely associated with lipid synthesis, oxidative stress, and gut microbiota balance. Moreover, PXR plays dual roles in various cancers, including hepatocellular carcinoma, colorectal cancer, and breast cancer. Currently, PXR-targeted strategies, such as small molecule agonists and antagonists, represent promising therapeutic avenues for treating metabolic diseases and cancer. This review comprehensively summarizes the structural features, signaling pathways, and gene regulatory functions of PXR, as well as its role in metabolic diseases and cancer, providing insights into its therapeutic potential and future drug development challenges. Full article
(This article belongs to the Section Molecular Endocrinology and Metabolism)
Show Figures

Figure 1

21 pages, 967 KiB  
Review
Recent Advances in the Application of Cucurbitacin B as an Anticancer Agent
by Dongge Yin, Hongyue Chen, Shuting Lin, Yufei Sun, Xiaohong Jing, Rongrong Chang, Yang Feng, Xiaoxv Dong, Changhai Qu, Jian Ni and Xingbin Yin
Int. J. Mol. Sci. 2025, 26(16), 8003; https://doi.org/10.3390/ijms26168003 - 19 Aug 2025
Viewed by 296
Abstract
Cucurbitacin B (CuB), a tetracyclic triterpenoid compound isolated from Cucurbitaceae plants, exhibits inhibitory effects on various tumor cells (e.g., liver, gastric, and colorectal cancer cells). Since the 1970s–1980s, cucurbitacin tablets containing CuB have been used as an adjuvant therapy for chronic hepatitis and [...] Read more.
Cucurbitacin B (CuB), a tetracyclic triterpenoid compound isolated from Cucurbitaceae plants, exhibits inhibitory effects on various tumor cells (e.g., liver, gastric, and colorectal cancer cells). Since the 1970s–1980s, cucurbitacin tablets containing CuB have been used as an adjuvant therapy for chronic hepatitis and primary liver cancer. CuB exerts anticancer effects through multiple mechanisms: inducing apoptosis, cell cycle arrest (G2/M or S phase), autophagy, and cytoskeleton disruption; inhibiting migration, invasion, and angiogenesis (via VEGF/FAK/MMP-9 and Wnt/β-catenin pathways); regulating metabolic reprogramming and immune responses; inducing pyroptosis, ferroptosis, and epigenetic changes; and reversing tumor drug resistance. These effects are associated with signaling pathways like JAK/STAT, PI3K/Akt/mTOR, and FOXM1-KIF20A. To improve its application potential, strategies such as structural modification (e.g., NO donor conjugation), combination therapy (with gemcitabine or cisplatin), and nanomaterial-based delivery (e.g., liposomes and exosome-mimicking nanoparticles) have been developed to enhance efficacy, reduce toxicity, and improve bioavailability. CuB shows broad-spectrum anticancer activity, but further research is needed to clarify the mechanisms underlying its cell-specific sensitivity and interactions with the immune system. This review systematically summarizes the physicochemical properties, anticancer mechanisms, and strategies for applying CuB and suggests future research directions, providing references for scientific research and clinical translation. Full article
(This article belongs to the Section Molecular Plant Sciences)
Show Figures

Graphical abstract

15 pages, 1929 KiB  
Article
Direct oHSV Infection Induces DC Maturation and a Tumor Therapeutic Response
by Doyeon Kim, Michael Kelly, Jack Hedberg, Alexia K. Martin, Ilse Hernandez-Aguirre, Yeaseul Kim, Lily R. Cain, Ravi Dhital and Kevin A. Cassady
Viruses 2025, 17(8), 1134; https://doi.org/10.3390/v17081134 - 19 Aug 2025
Viewed by 259
Abstract
Oncolytic herpes simplex virus (oHSV) is a promising cancer immunotherapy that induces tumor cell lysis and stimulates anti-tumor immunity. Our previous single-cell RNA sequencing analysis of oHSV-treated medulloblastoma tumors revealed expansion and activation of tumor-infiltrating dendritic cells (DCs), and direct oHSV infection of [...] Read more.
Oncolytic herpes simplex virus (oHSV) is a promising cancer immunotherapy that induces tumor cell lysis and stimulates anti-tumor immunity. Our previous single-cell RNA sequencing analysis of oHSV-treated medulloblastoma tumors revealed expansion and activation of tumor-infiltrating dendritic cells (DCs), and direct oHSV infection of DCs within the brain. While the therapeutic effects of oHSVs have been primarily attributed to tumor cell infection, we hypothesize that direct infection of DCs also contributes to therapeutic efficacy by promoting DC maturation and immune activation. Although the oHSV infection in DCs was abortive, it led to increased expression of major histocompatibility complex (MHC) class I/II and co-stimulatory molecules. oHSV-infected DCs activated naïve CD4+ and CD8+ T cells, inducing expression of CD69 and CD25. These primed T cells exhibited enhanced cytotoxicity against CT-2A glioma cells. Adoptive transfer of oHSV-infected DCs via subcutaneous injection near inguinal lymph nodes delayed tumor growth in a syngeneic CT-2A glioma model, independent of tumor viral replication and lysis. Mechanistically, our in vitro studies demonstrate that oHSV can directly infect and functionally activate DCs, enabling them to prime effective anti-tumor T cell responses. This study highlights the anti-tumor potential of leveraging oHSV-infected DCs to augment viroimmunotherapy as a cancer therapeutic. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

15 pages, 1012 KiB  
Review
Exploring the Therapeutic Potential of Bovine Colostrum for Cancer Therapies
by Yalçın Mert Yalçıntaş, Mikhael Bechelany and Sercan Karav
Int. J. Mol. Sci. 2025, 26(16), 7936; https://doi.org/10.3390/ijms26167936 - 17 Aug 2025
Viewed by 397
Abstract
Colostrum is a nutrient-rich fluid secreted by mammals shortly after birth, primarily to provide passive immunity and support early immune development in newborns. Among its various sources, bovine colostrum is the most widely used supplement due to its high bioavailability, safety profile, and [...] Read more.
Colostrum is a nutrient-rich fluid secreted by mammals shortly after birth, primarily to provide passive immunity and support early immune development in newborns. Among its various sources, bovine colostrum is the most widely used supplement due to its high bioavailability, safety profile, and clinically supported health benefits. Rich in immunoglobulins, lactoferrin, growth factors, and antimicrobial peptides, bovine colostrum exhibits diverse biological activities that extend beyond neonatal health. Recently, the rising prevalence of cancer—driven by environmental stressors such as radiation, processed foods, and chronic inflammation, as well as non-environmental hereditary factors including germline mutations, family history, and epigenetic inheritance—has fueled interest in natural adjunctive therapies. Scientific studies have explored the anticancer potential of bovine colostrum, highlighting its ability to modulate immune responses, inhibit tumor growth, induce apoptosis in cancer cells, and reduce inflammation. Key components including lactoferrin and proline-rich peptides have been identified as contributors to these effects. Additionally, bovine colostrum may help reduce the side effects of standard cancer treatments, such as mouth sores from chemotherapy or weakened immune systems, by helping to heal tissues and boost the body’s defenses. While large-scale clinical studies are still needed, current findings suggest that bovine colostrum holds promise as a supportive element in integrative cancer care. In conclusion, bovine colostrum represents a safe, bioactive-rich natural supplement with multifaceted therapeutic potential, particularly in oncology, owing to its key components such as lactoferrin, immunoglobulins, growth factors (e.g., IGF-1, TGF-β), and proline-rich polypeptides (PRPs), which contribute to its immunomodulatory, anti-inflammatory, and potential anticancer effects. Ongoing and future research will be crucial to fully understand its mechanisms of action and establish its role in evidence-based cancer prevention and treatment strategies. Full article
Show Figures

Figure 1

34 pages, 1707 KiB  
Review
Mimicking Gastric Cancer Collagen Reorganization with Decellularized ECM-Based Scaffolds
by Néstor Corro, Sebastián Alarcón, Ángel Astroza, Roxana González-Stegmaier and Carolina Añazco
Biology 2025, 14(8), 1067; https://doi.org/10.3390/biology14081067 - 16 Aug 2025
Viewed by 349
Abstract
The tumor microenvironment (TME) has a substantial impact on the progression of gastric cancer. Collagen, the most abundant protein in the extracellular matrix (ECM), forms a dense physical barrier that regulates anti-tumor immunity in the TME. It is a significant regulator of the [...] Read more.
The tumor microenvironment (TME) has a substantial impact on the progression of gastric cancer. Collagen, the most abundant protein in the extracellular matrix (ECM), forms a dense physical barrier that regulates anti-tumor immunity in the TME. It is a significant regulator of the signaling pathways of cancer cells, which are responsible for migration, proliferation, and metabolism. ECM proteins, particularly remodeling enzymes and collagens, can be modified to increase stiffness and alter the mechanical properties of the stroma. This, in turn, increases the invasive potential of tumor cells and resistance to immunotherapy. Given the dynamic nature of collagen, novel therapeutic strategies have emerged that target both collagen biosynthesis and degradation, processes that are essential for addressing ECM stiffening. This review delineates the upregulation of the expression and deposition of collagen, as well as the biological functions, assembly, and reorganization that contribute to the dissemination of this aggressive malignancy. Furthermore, the review emphasizes the importance of creating 3D in vitro models that incorporate innovative biomaterials that avoid the difficulties of traditional 2D culture in accurately simulating real-world conditions that effectively replicate the distinctive collagen microenvironment. Ultimately, it investigates the use of decellularized ECM-derived biomaterials as tumor models that are designed to precisely replicate the mechanisms associated with the progression of stomach cancer. Full article
(This article belongs to the Special Issue Tumor Biomechanics and Mechanobiology)
Show Figures

Graphical abstract

25 pages, 4622 KiB  
Review
Immunological Landscape and Molecular Therapeutic Targets of the Tumor Microenvironment in Hepatocellular Carcinoma
by Yusra Zarlashat, Abdul Ghaffar, Flora Guerra and Anna Picca
Int. J. Mol. Sci. 2025, 26(16), 7836; https://doi.org/10.3390/ijms26167836 - 13 Aug 2025
Viewed by 441
Abstract
Hepatocellular carcinoma (HCC) is the most common liver cancer, with poor survival rates in advanced stages due to late diagnosis, tumor heterogeneity, and therapy resistance. The tumor microenvironment (TME) in HCC has a crucial role in tumor progression, characterized by a complex interaction [...] Read more.
Hepatocellular carcinoma (HCC) is the most common liver cancer, with poor survival rates in advanced stages due to late diagnosis, tumor heterogeneity, and therapy resistance. The tumor microenvironment (TME) in HCC has a crucial role in tumor progression, characterized by a complex interaction of immune cells, stromal components, and immunosuppressive signaling pathways. Chronic inflammation driven by viral infections, metabolic dysfunction, and alcohol consumption triggers an immunosuppressive TME, promoting immune evasion and tumor growth. Immune cell populations, such as myeloid-derived suppressor cells, regulatory T cells, and tumor-associated macrophages, contribute to immunosuppression, while cytotoxic T lymphocytes and natural killer cells exert anti-tumor effects. Recent advances in immunotherapy, mainly immune checkpoint inhibitors (ICIs) targeting programmed death-ligand 1 and programmed cell death protein 1 and cytotoxic T-lymphocyte-associated protein 4, have revolutionized HCC treatment, though response rates remain limited. Combined therapies using tyrosine kinase inhibitors, anti-angiogenic agents, and ICIs improve patient outcomes. This review discusses the immunological mechanisms contributing to HCC progression, the role of immune cell subsets in tumor evasion, and therapeutic interventions, from conventional treatments to advanced immunotherapies. Ongoing clinical trials, barriers to effective treatment, and future directions to enhance HCC management and patient survival will also be overviewed. Full article
Show Figures

Figure 1

13 pages, 1252 KiB  
Article
Prognostic Impact of Gastrointestinal Immune-Related Adverse Events Depends on Nutritional Status in Cancer Patients Treated with Immune Checkpoint Inhibitors
by Shoichiro Hirata, Yoshiyasu Kono, Emi Tanaka, Masahiko Sue, Yasuto Takeuchi, Tomoki Yoshikawa, Yoshie Maki, Tomohiro Kamio, Daisuke Kametaka, Katsunori Matsueda, Chihiro Sakaguchi, Kenta Hamada, Masaya Iwamuro, Seiji Kawano, Yoshiro Kawahara and Motoyuki Otsuka
Cancers 2025, 17(16), 2634; https://doi.org/10.3390/cancers17162634 - 12 Aug 2025
Viewed by 490
Abstract
Background: Gastrointestinal immune-related adverse events (GI-irAEs) are recognized complications of immune checkpoint inhibitors (ICIs), but their prognostic relevance and associated risk factors remain unclear. This study aimed to assess whether baseline nutritional status, measured using the prognostic nutritional index (PNI), modifies the prognostic [...] Read more.
Background: Gastrointestinal immune-related adverse events (GI-irAEs) are recognized complications of immune checkpoint inhibitors (ICIs), but their prognostic relevance and associated risk factors remain unclear. This study aimed to assess whether baseline nutritional status, measured using the prognostic nutritional index (PNI), modifies the prognostic impact of GI-irAEs, and to identify clinical factors associated with their occurrence. Methods: We retrospectively analyzed 1104 cancer patients treated with ICIs at a single institution. GI-irAEs were defined as gastrointestinal symptoms requiring clinical intervention. Patients were stratified by irAE type and PNI (≥40 vs. <40), and differences in survival and treatment response were evaluated. Potential risk factors for developing GI-irAEs were also examined. Results: GI-irAEs occurred in 2.7% of patients and were associated with prolonged overall survival (median: 28.7 vs. 14.0 months) among those with PNI ≥ 40. This survival advantage was not observed in patients with PNI < 40. The PNI-dependent prognostic pattern was specific to GI-irAEs and not observed for non-GI irAEs. Similar trends were confirmed in 4- and 8-week landmark analyses. Differences in objective response rate and disease control rate by PNI status were most pronounced in patients with GI-irAEs. The use of anti-CTLA-4 antibodies was significantly associated with GI-irAE development (odds ratio 4.24; 95% confidence interval 1.73–10.39). Conclusions: GI-irAEs appear to confer a survival benefit primarily in patients with preserved nutritional status. PNI may serve as a useful tool to contextualize the clinical relevance of GI-irAEs and help identify patients most likely to benefit from immune activation during ICI therapy. Full article
(This article belongs to the Special Issue Advances in the Treatment of Gastrointestinal (GI) Cancers)
Show Figures

Figure 1

17 pages, 10690 KiB  
Article
Tumor Microenvironment Specifically Regulated Nano Chemoamplifier for Chemosensitization and Activation of Anti-Tumor Immune Response by Coordinating Intracellular Magnesium Overload
by Chao Liu, Gaofei Huang, Lu Zhu, Shasha Li, Kun Yang, Nuernisha Alifu and Yingni Duan
Pharmaceutics 2025, 17(8), 1034; https://doi.org/10.3390/pharmaceutics17081034 - 9 Aug 2025
Viewed by 365
Abstract
Background and objectives: Chemotherapy is an established treatment modality for breast cancer; however, it is impaired by issues such as highly refractory chemoresistance and significant side effects. Magnesium ions (Mg2+), inorganic metal ions with anti-tumor bioactivity, sensitize cancer cells to chemotherapy [...] Read more.
Background and objectives: Chemotherapy is an established treatment modality for breast cancer; however, it is impaired by issues such as highly refractory chemoresistance and significant side effects. Magnesium ions (Mg2+), inorganic metal ions with anti-tumor bioactivity, sensitize cancer cells to chemotherapy by depressing P-glycoprotein (P-gp) expression. Moreover, Mg2+ functions as an immunoadjuvant to potentiate anti-tumor immune responses, while excessive Mg2+ can induce marked tumor cell apoptosis. Methods: To enable Mg2+ to serve as a chemotherapeutic adjuvant for enhanced treatment efficacy, a Trojan horse-like chemoamplifier, denoted as MMSN@Dox, endowed with tumor microenvironment (TME) responsiveness and capable of achieving chemotherapy sensitization and anti-tumor immune activation, was constructed to enhance the efficacy of breast cancer treatment. Leveraging Mg2+-enabled TME-responsive degradability of the chemoamplifier, density functional theory (DFT) simulations were conducted to elucidate carrier structural dynamics. Results: Under stimulation of TME, the chemoamplifier decomposes, accompanied by a substantial release of chemotherapeutic agents and metal ions. Excessive Mg2+ induces significant tumor cell apoptosis by triggering mitochondrial dysfunction and generating reactive oxygen species (ROS), and reinforces chemotherapy sensitivity by depressing P-gp expression. Furthermore, MMSN@Dox weakens the stemness of tumor cells, further enhancing chemotherapy. The remarkable tumor-killing capability of chemoamplifier MMSN@Dox led to a remarkable immunogenic cell death (ICD) effect. Combined with the regulatory function of Mg2+ on T cells, it ultimately activates anti-tumor immune responses and achieves exceptional anti-tumor performance in both in vitro and in vivo models. Conclusions: This approach, leveraging Mg2+ to enhance chemotherapy efficacy, establishes a new paradigm for overcoming chemotherapy resistance and offers a novel strategic avenue for advancing nanomedicine in breast cancer treatment. Full article
(This article belongs to the Section Nanomedicine and Nanotechnology)
Show Figures

Figure 1

30 pages, 2188 KiB  
Review
A Comprehensive Review of Fortification, Bioavailability, and Health Benefits of Folate
by Jiarun Li, Hanying Duan, Hosahalli Ramaswamy and Chao Wang
Int. J. Mol. Sci. 2025, 26(16), 7703; https://doi.org/10.3390/ijms26167703 - 9 Aug 2025
Viewed by 762
Abstract
Folate is an essential vitamin involved in one-carbon metabolism. It can be acquired from many food sources or in synthetic form. A wide range of processing methods have been studied to improve the bioaccessibility and bioavailability of folate in foods, yet this is [...] Read more.
Folate is an essential vitamin involved in one-carbon metabolism. It can be acquired from many food sources or in synthetic form. A wide range of processing methods have been studied to improve the bioaccessibility and bioavailability of folate in foods, yet this is often accompanied by a decrease in stability. Encapsulation technology has emerged as an effective solution for protecting folate from degradation and liberation while also improving its bioavailability. Folate deficiency is a prevalent phenomenon worldwide, particularly in underprivileged countries, leading to various health problems, such as neural tube defects. Thus, folate was fortified through both exogenous addition and biofortification. Gene editing technology, especially CRISPR-Cas9, has great promise in this field when compared to transgenic engineering, because transgenic engineering may pose safety concerns and environmental risks. While ongoing research has identified additional potential effects of folate, the dosage and duration remain important factors to consider for optimal health outcomes. The mechanisms of how folate promotes the production of neurotransmitters associated with the gut microbiota–brain axis and reduces depression are not well understood. In addition to folate alone, there may be synergistic effects of combined supplementation of folate and other nutrients or medications, but this is not yet fully clarified and requires further examination. This review summarizes the food sources, enrichment, bioaccessibility, and bioavailability of folate. Furthermore, the health benefits of folate, including neural tube protection, cardiovascular protection, neuroprotection, anti-cancer, immune response augmentation, and gut homeostasis maintenance, with their potential bioactivity mechanisms, are discussed. Full article
Show Figures

Graphical abstract

15 pages, 3491 KiB  
Article
PARP Inhibition Shifts Murine Myeloid Cells Toward a More Tolerogenic Profile In Vivo
by Jose R. Pittaluga-Villarreal, Casey M. Daniels, Tara Capece, Pauline R. Kaplan, Martin Meier-Schellersheim and Aleksandra Nita-Lazar
Biomolecules 2025, 15(8), 1149; https://doi.org/10.3390/biom15081149 - 9 Aug 2025
Viewed by 446
Abstract
The human Poly ADP-ribose Polymerase (PARP) family comprises 17 enzymes responsible for the transfer of ADP-ribose to proteins, forming poly- or mono-ADP-ribosylation. This post-translational modification regulates DNA repair and programmed cell death, processes affecting cancer biology. PARP inhibitors, including the FDA-approved olaparib, are [...] Read more.
The human Poly ADP-ribose Polymerase (PARP) family comprises 17 enzymes responsible for the transfer of ADP-ribose to proteins, forming poly- or mono-ADP-ribosylation. This post-translational modification regulates DNA repair and programmed cell death, processes affecting cancer biology. PARP inhibitors, including the FDA-approved olaparib, are used to treat BRCA-dependent breast and ovarian cancers. Although therapies with use of PARP inhibitors are showing clinical success, their effects on the immune system remain understudied. Prior work has shown that PARP inhibition can modulate inflammatory responses and alter innate immunity. In this study, we evaluated the immunomodulatory effects of olaparib on myeloid cells in vivo, focusing on bone marrow and spleen. Olaparib treatment altered the composition and activation state of dendritic cells, neutrophils, and macrophages. In the bone marrow, olaparib increased the proportion of cDC2 population, mature neutrophils and inflammatory macrophages expressing CD80. In contrast, splenic myeloid cells exhibited enhanced expression of markers associated with tolerogenic phenotypes, including CD206 and CD124 in neutrophils and macrophages. The spleen also showed an increase in immature monocyte-derived dendritic cells (CD206+) and a bias toward the cDC2 subset. These findings indicate that PARP inhibition can induce short-term phenotypic remodeling of myeloid cell populations, promoting a more immunoregulatory profile, especially in the spleen. These changes may contribute to an altered immune landscape with implications for anti-tumor immunity. Full article
(This article belongs to the Special Issue PARPs in Cell Death and PARP Inhibitors in Cancers: 2nd Edition)
Show Figures

Figure 1

29 pages, 2716 KiB  
Review
Dendritic Cells and Their Crucial Role in Modulating Innate Lymphoid Cells for Treating and Preventing Infectious Diseases
by Yeganeh Mehrani, Solmaz Morovati, Fatemeh Keivan, Tahmineh Tajik, Diba Forouzanpour, Sina Shojaei, Byram W. Bridle and Khalil Karimi
Pathogens 2025, 14(8), 794; https://doi.org/10.3390/pathogens14080794 - 8 Aug 2025
Viewed by 343
Abstract
Two key players in the immune system, dendritic cells (DCs) and innate lymphoid cells (ILCs), interact in a crucial way to fight infectious diseases. DCs play a key role in recognizing pathogens, and ILCs respond to cytokines released by DCs. This response triggers [...] Read more.
Two key players in the immune system, dendritic cells (DCs) and innate lymphoid cells (ILCs), interact in a crucial way to fight infectious diseases. DCs play a key role in recognizing pathogens, and ILCs respond to cytokines released by DCs. This response triggers the production of specific effector cytokines that help control pathogens and maintain the body’s barrier integrity. DCs have various receptors, including Toll-like receptors (TLRs), that detect microbial components and trigger immune responses. Likewise, ILCs act as essential initial responders in the immune system in viral, bacterial, and parasitic infections. Successfully managing diseases caused by pathogens mainly depends on the combined actions of DCs and ILCs, which work to suppress and eliminate pathogens. DCs also play a crucial role in activating innate and adaptive immune cell subsets, including ILCs. Furthermore, the use of DCs in developing vaccines and immunotherapy for cancers, along with the dedication of many researchers to improve immune responses through DCs, has increased interest in the potential of DC therapies for treating and preventing infectious diseases. This review examines approaches that may enhance DC vaccines and boost anti-infection immune responses by fostering better interactions of DCs with ILCs. Full article
Show Figures

Figure 1

21 pages, 1727 KiB  
Review
Immune Evasion in Head and Neck Squamous Cell Carcinoma: Roles of Cancer-Associated Fibroblasts, Immune Checkpoints, and TP53 Mutations in the Tumor Microenvironment
by Chung-Che Tsai, Yi-Chiung Hsu, Tin-Yi Chu, Po-Chih Hsu and Chan-Yen Kuo
Cancers 2025, 17(15), 2590; https://doi.org/10.3390/cancers17152590 - 7 Aug 2025
Viewed by 590
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a highly aggressive malignancy characterized by complex interactions within the tumor microenvironment (TME) that facilitate immune evasion and tumor progression. The TME consists of diverse cellular components, including cancer-associated fibroblasts, immune and endothelial cells, and [...] Read more.
Head and neck squamous cell carcinoma (HNSCC) is a highly aggressive malignancy characterized by complex interactions within the tumor microenvironment (TME) that facilitate immune evasion and tumor progression. The TME consists of diverse cellular components, including cancer-associated fibroblasts, immune and endothelial cells, and extracellular matrix elements, that collectively modulate tumor growth, metastasis, and resistance to therapy. Immune evasion in HNSCC is orchestrated through multiple mechanisms, including the suppression of cytotoxic T lymphocytes, recruitment of immunosuppressive cells, such as regulatory T and myeloid-derived suppressor cells, and upregulation of immune checkpoint molecules (e.g., PD-1/PD-L1 and CTLA-4). Natural killer (NK) cells, which play a crucial role in anti-tumor immunity, are often dysfunctional within the HNSCC TME due to inhibitory signaling and metabolic constraints. Additionally, endothelial cells contribute to tumor angiogenesis and immune suppression, further exacerbating disease progression. Recent advancements in immunotherapy, particularly immune checkpoint inhibitors and NK cell-based strategies, have shown promise in restoring anti-tumor immunity. Moreover, TP53 mutations, frequently observed in HNSCC, influence tumor behavior and therapeutic responses, highlighting the need for personalized treatment approaches. This review provides a comprehensive analysis of the molecular and cellular mechanisms governing immune evasion in HNSCC with a focus on novel therapeutic strategies aimed at improving patient outcomes. Full article
(This article belongs to the Special Issue Oral Cancer: Prevention and Early Detection (2nd Edition))
Show Figures

Figure 1

17 pages, 2609 KiB  
Article
Residual Tumor Resection After Anti-PD-1 Therapy: A Promising Treatment Strategy for Overcoming Immune Evasive Phenotype Induced by Anti-PD-1 Therapy in Gastric Cancer
by Hajime Matsuida, Kosaku Mimura, Shotaro Nakajima, Katsuharu Saito, Sohei Hayashishita, Chiaki Takiguchi, Azuma Nirei, Tomohiro Kikuchi, Hiroyuki Hanayama, Hirokazu Okayama, Motonobu Saito, Tomoyuki Momma, Zenichiro Saze and Koji Kono
Cells 2025, 14(15), 1212; https://doi.org/10.3390/cells14151212 - 6 Aug 2025
Viewed by 400
Abstract
Background: Anti-programmed death 1 receptor (PD-1) therapy is a promising treatment strategy for patients with unresectable advanced or recurrent gastric/gastroesophageal junction (G/GEJ) cancer. However, its response rate and survival benefits are still limited; an immunological analysis of the residual tumor after anti-PD-1 therapy [...] Read more.
Background: Anti-programmed death 1 receptor (PD-1) therapy is a promising treatment strategy for patients with unresectable advanced or recurrent gastric/gastroesophageal junction (G/GEJ) cancer. However, its response rate and survival benefits are still limited; an immunological analysis of the residual tumor after anti-PD-1 therapy would be important. Methods: We evaluated the clinical efficacy of tumor resection (TR) after chemotherapy or anti-PD-1 therapy in patients with unresectable advanced or recurrent G/GEJ cancer and analyzed the immune status of tumor microenvironment (TME) by immunohistochemistry using their surgically resected specimens. Results: Patients treated with TR after anti-PD-1 therapy had significantly longer survival compared to those treated with chemotherapy and anti-PD-1 therapy alone. Expression of human leukocyte antigen (HLA) class I and major histocompatibility complex (MHC) class II on tumor cells was markedly downregulated after anti-PD-1 therapy compared to chemotherapy. Furthermore, the downregulation of HLA class I may be associated with the activation of transforming growth factor-β signaling pathway in the TME. Conclusions: Immune escape from cytotoxic T lymphocytes may be induced in the TME in patients with unresectable advanced or recurrent G/GEJ cancer after anti-PD-1 therapy due to the downregulation of HLA class I and MHC class II expression on tumor cells. TR may be a promising treatment strategy for these patients when TR is feasible after anti-PD-1 therapy. Full article
Show Figures

Figure 1

Back to TopTop