1
|
Zhang M, Qiu H, Han Z, Ma Y, Hou J, Yuan J, Jia H, Zhou M, Lu H, Wu Y. Topical transdermal administration of lenalidomide nanosuspensions-based hydrogels against melanoma: In vitro and in vivo studies. Int J Pharm X 2025; 9:100316. [PMID: 39898009 PMCID: PMC11787432 DOI: 10.1016/j.ijpx.2025.100316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 02/04/2025] Open
Abstract
Percutaneous neoadjuvant therapy has proven effective in diminishing tumor size and the surgical intervention area, which couldeffectively mitigate the risk of tumor recurrence and enhance immunotherapy efficacy. Lenalidomide, an approved medication orally used to treat myeloma, was loaded into nanosuspensions-based hydrogels (Len-NBHs) for transdermal administration as a percutaneous neoadjuvant therapy. This study was designed to investigate the inhibitory effect and mechanism of Len-NBHs on melanoma. Network pharmacology and transcriptomic analyses identified key targets and signaling pathways. The effects of lenalidomide on melanoma were further verified through Western blotting, immunohistochemistry, immunofluorescence, and quantitative real-time polymerase chain reaction,using both in vitro cell experiments and in vivo melanoma mouse models. Lenalidomide could induce melanoma cells apoptosis, disrupt cell cycle progression, impede cell migration and invasion, and modify tumor microenvironment (TME). Mechanistically, lenalidomide reversed the abnormal activation of the PI3K-AKT signaling pathway and the overexpression of CD93, while also recruiting CD8+ T cells, CD4+ T cells, and dendritic cells to infiltrate the tumor site. Transdermal administration of Len-NBHs represents a promising adjuvant therapy for the treatment of malignant melanoma. Preoperative administration of Len-NBHs can inhibit the outward spread of melanoma, reduce tumor size, thereby decreasing the surgical excision area and improving patient survival rates and prognosis.
Collapse
Affiliation(s)
| | | | - Zheyi Han
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Yazhong Ma
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Jingjing Hou
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Jingwei Yuan
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Haiyan Jia
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Menglu Zhou
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Hongjie Lu
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| | - Yan Wu
- Air Force Medical Center, PLA, Air Force Medical University, Beijing, China
| |
Collapse
|
2
|
Ramzy A, Abdel-Halim M, Manie T, Elemam NM, Mansour S, Youness RA, Sebak A. In-vitro immune-modulation of triple-negative breast cancer through targeting miR-30a-5p/MALAT1 axis using nano-PDT combinational approach. Transl Oncol 2025; 55:102365. [PMID: 40132387 PMCID: PMC11984585 DOI: 10.1016/j.tranon.2025.102365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Triple negative breast cancer (TNBC) is an immunogenic tumor; however, its tumor immune microenvironment (TIME) is densely packed with immune suppressive cytokines and immune checkpoints. The immune-suppressive features of TNBC TIME represent a considerable obstacle to any immunotherapeutic approach. The objective of this study was to develop a multimodal in-vitro strategy to manipulate the TNBC TIME and enhance patients' outcomes by employing carefully tailored hybrid chitosan-lipid Nanoparticles (CLNPs), metformin and chlorin e6 (Ce-6)-mediated PDT, alone or combined. Special focus is directed towards evaluation of the role of the selected treatment agents on the non-coding RNAs (ncRNAs) involved in tuning the immuno-oncogenic profile of TNBC, for instance, the miR-30a-5p/MALAT1 network. METHODS This study enrolled 30 BC patients. CLNPs and ce-6-loaded CLNPs with different physicochemical features were synthesized and optimized using ionotropic gelation. The intracellular concentration and effects on MDA-MB-231 cellular viability were investigated. UHPLC was used to quantify ce-6. MDA-MB-231 cells were transfected with miR-30a-5p oligonucleotides and MALAT1 siRNAs using lipofection to investigate the interaction between MIF, PD-L1, TNF-α, IL-10, and the miR-30a-5p/MALAT1 ceRNA network. qRT-PCR was used to evaluate IL-10, TNF-α, and MIF expression levels, whereas flow cytometry was used for PD-L1. RESULTS Immunophenotyping of BC biopsies revealed significantly elevated levels of immunosuppressive markers, including IL-10, TNF-α, PD-L1, and MIF in BC biopsies compared to its normal counterparts. Upon patient stratification, it was shown that MIF and IL-10 are upregulated in TNBC patients compared to non-TNBC patients. Nonetheless, immune suppressive biomarkers expression investigated in the current study was generally correlated with signs of poor prognosis. CLNPs with mean particle size ranging from 50-150 nm were obtained. CLNPs exhibited different patterns of intracellular uptake, cytotoxicity and modulation of the immunosuppressive markers based on their physicochemical properties and composition. In particular, CLNP4 in-vitro effectively reduced IL-10, TNF-α, MIF, and PD-L1. Loading of Ce-6 into CLNP4 (Ce6-CLNPs) improved the in-vitro cytotoxic effects via PDT. In addition, PDT with Ce6-CLNP4 enhanced the expression of tumor-suppressive miR-30a-5p and decreased oncogenic lncRNA MALAT1 expression in MDA-MB-231 cells, suggesting a potential for modulating the TNBC immuno-oncogenic profile. CONCLUSION This study demonstrated that CLNPs and Ce-6-mediated PDT can modulate several key immunosuppressive factors and the miR-30a-5p/MALAT1 axis in TNBC cells. These findings provide a rationale for further in-vivo investigation of this multimodal therapeutic strategy.
Collapse
Affiliation(s)
- Asmaa Ramzy
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, New Cairo 11835, Egypt
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy & Biotechnology, The German University in Cairo, New Cairo 11835, Egypt
| | - Tamer Manie
- Department of Breast Surgery, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Noha M Elemam
- Clinical Sciences Department, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Samar Mansour
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, New Cairo 11835, Egypt; Faculty of Pharmaceutical Engineering, German International University (GIU), New Administrative Capital, Cairo 11835, Egypt
| | - Rana A Youness
- Department of Molecular Biology and Biochemistry, Molecular Genetics Research Team (MGRT), Faculty of Biotechnology, German International University (GIU), New Administrative Capital, Cairo 11835, Egypt.
| | - Aya Sebak
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo, New Cairo 11835, Egypt.
| |
Collapse
|
3
|
Hema Shree K, Gayathri R, Veeraraghavan VP, Ramani P, Ramadoss R, Yuwanati M. Gold nanoparticle enhanced TNFα antibody interface using saliva for predicting prognosis in OSCC. Arch Oral Biol 2025; 173:106196. [PMID: 39961150 DOI: 10.1016/j.archoralbio.2025.106196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 04/13/2025]
Abstract
OBJECTIVES To assess the efficacy of the AuNP-enhanced ELISA in the detection of salivary TNF-α in OSCC and evaluate its predictive value for survival. DESIGN A longitudinal study was conducted on 40 OSCC patients and 10 healthy controls. Saliva was collected at a regular interval and TNF alpha levels were measured using ELISA and AuNP-enhanced ELISA. Descriptive statistics were carried out for demographic, clinical, and biomarker data. The efficacy of ELISA and AuNP-enhanced ELISA was estimated and compared using the ROC curve. Kaplan-Meier survival analysis and Cox proportional hazards models were used to assess the survival outcome. A neural network model was performed to estimate TNF-α levels over time in OSCC patients. RESULTS OSCC patients (ELISA is 47.52 ± 20.23 and Gold nano-enhanced ELISA is 57.63 ± 24.99) exhibited significantly elevated TNFα levels compared to controls (ELISA is 10.13 ± 3.07and Gold nano-enhanced ELISA is 12.07 ± 3.66). Gold nano-enhanced ELISA (AUC of 0.995) demonstrated superior sensitivity than ELISA (AUC of 0.986), while Gold nano-enhanced ELISA achieves an even higher in detecting elevated TNFα levels. Kaplan-Meier analysis shows that Gold nano-enhanced ELISA outperforms ELISA in capturing survival trends, with better survival for TNF-α levels above the cutoff at 9 months (70 % vs. 60 %) and 24 months (40 % vs. 0 %). The neural network model poorly predicted TNF- levels over the period (AUC = 0.447). CONCLUSIONS The gold nano-enhanced TNFα detection method is effective in detecting TNFα levels between OSCC patients and controls, demonstrating superior sensitivity in identifying survival trends over time compared to traditional ELISA.
Collapse
Affiliation(s)
- K Hema Shree
- Department of Oral and Maxillofacial Pathology, Saveetha Dental College and Hospitals, Saveetha Institute Medical and Technical Science, Saveetha University, Chennai, India.
| | - R Gayathri
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute Medical and Technical Science, Saveetha University, Chennai, India.
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute Medical and Technical Science, Saveetha University, Chennai, India.
| | - Pratibha Ramani
- Department of Oral and Maxillofacial Pathology, Saveetha Dental College and Hospitals, Saveetha Institute Medical and Technical Science, Saveetha University, Chennai, India.
| | - Ramya Ramadoss
- Department of Oral Biology, Saveetha Dental College and Hospitals, Saveetha Institute Medical and Technical Science, Saveetha University, Chennai, India.
| | - Monal Yuwanati
- Department of Oral and Maxillofacial Pathology, Saveetha Dental College and Hospitals, Saveetha Institute Medical and Technical Science, Saveetha University, Chennai, India.
| |
Collapse
|
4
|
Pele KG, Calderón-Villalba A, Amaveda H, Mora M, Zhang-Zhou J, Pérez MÁ, García-Aznar JM, Alamán-Díez P, García-Gareta E. Novel hydrogel-based cancer-on-a-chip models for growth of 3D multi-cellular structures and investigation of early angiogenesis in pancreatic ductal adenocarcinoma. Colloids Surf B Biointerfaces 2025; 253:114736. [PMID: 40315572 DOI: 10.1016/j.colsurfb.2025.114736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 03/27/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
Cancer-on-a-chip models have enormous potential for the study of tumour development events. Here, we investigated hydrogels of egg white (EW) and gelatin for growth of 3D multi-cellular structures and investigation of early angiogenesis inside microfluidic devices. We focused on pancreatic ductal adenocarcinoma (PDAC), a devastating gastrointestinal malignancy. EW/gelatin hydrogels were stiffer and showed porous globular structures compared to the fibrous network of collagen I molecules. PANC-1 cells preferentially formed significantly larger spheroids in collagen I than in EW/gelatin hydrogels, whilst cell aggregates in the shape of grape-like clusters were significantly larger and more abundant in EW/gelatin. Cells inside the aggregates showed active cell unions, secreted matrix, and formed active unions with the surrounding EW/gelatin hydrogel. Early stages of PDAC were recreated by co-culture of two different microenvironments, one for PANC-1 and another one for fibroblasts, for investigating the secretion of soluble angiogenic factors, which depended on the role of each factor in the angiogenic and tumorigenic processes. Overall, cancer cell proliferation and establishment of a tumour vasculature were favoured. This study demonstrates the importance of the microenvironment in tumour cells behaviour as well as the complex interplay between the different cells present in PDAC to establish a tumoural vasculature.
Collapse
Affiliation(s)
- Karinna Georgiana Pele
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Alejandro Calderón-Villalba
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Hippolyte Amaveda
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC and University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Mario Mora
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC and University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Jack Zhang-Zhou
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - María Ángeles Pérez
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain
| | - José Manuel García-Aznar
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain
| | - Pilar Alamán-Díez
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain
| | - Elena García-Gareta
- Multiscale in Mechanical & Biological Engineering Research Group, Aragon Institute of Engineering Research (I3A),School of Engineering & Architecture, University of Zaragoza, Zaragoza, Aragon 50018, Spain; Aragon Institute for Health Research (IIS Aragon), Miguel Servet University Hospital, Zaragoza, Aragon 50009, Spain; Division of Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, University College London, London NW3 2QG, United Kingdom.
| |
Collapse
|
5
|
Pati NB, Jin Y, Kumar S, Kyte JA, Ciosk R. Thermal potentiation improves IFN-γ production but not cytotoxicity in human CAR-T cells. BMC Res Notes 2025; 18:192. [PMID: 40270044 PMCID: PMC12020131 DOI: 10.1186/s13104-025-07249-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 04/09/2025] [Indexed: 04/25/2025] Open
Abstract
OBJECTIVE Body temperature plays an important role in cancer, with febrile temperature generally associated with improved cancer resistance. In murine models, this resistance has been linked to the cytotoxic T cells, whose differentiation into cancer-killing effector cells is poor at lower but robust at elevated temperatures. If conserved, temperature-mediated potentiation of patient-derived T cells could be implemented to improve the existing cancer treatments, including the chimeric antigen receptor T-cell therapy (CAR T-cell therapy). Here, we tested this possibility using human STEAP1 CAR-T cells developed to target prostate cancer. RESULTS In mice, transient exposure to febrile temperature (39-40 ºC) increases the production of IFN-γ and the cancer-killing ability of CD8 + T cells. Using a similar temperature treatment, we observed elevated levels of IFN-γ also in the human CAR-T cells. However, these cells displayed no improvement in their ability to kill cancer cells. Although we cannot discount the possibility that alternative protocols might lead to other outcomes, our findings suggest that incorporating thermal potentiation into existing protocols of CAR-T cell therapy may be more complicated than anticipated.
Collapse
Affiliation(s)
- Niladri Bhusan Pati
- Department of Biosciences, University of Oslo, Oslo, Norway
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Yixin Jin
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Suman Kumar
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Jon Amund Kyte
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway.
- Department of Clinical Cancer Research, Oslo University Hospital, Oslo, Norway.
- Faculty of Health Sciences, Oslo Metropolitan University, Oslo, Norway.
| | - Rafal Ciosk
- Department of Biosciences, University of Oslo, Oslo, Norway.
| |
Collapse
|
6
|
Singh AK, Kumar Pathak A, Kumar P, Kumar Singh A, Kumar Sah Gond M, Singh Negi R, Das R, Agrawal S, Kumar Mishra S, Tiwari KN. Effects of Asiatic acid on brain cancer by altering astrocytes and the AKT1-PRKCB signaling pathway: A genomic and network pharmacology perspective. Brain Res 2025; 1859:149652. [PMID: 40252893 DOI: 10.1016/j.brainres.2025.149652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 04/07/2025] [Accepted: 04/17/2025] [Indexed: 04/21/2025]
Abstract
The most common primary brain tumor, glioblastoma (GBM), currently has a dismal prognosis because of its fast growth and dissemination. Recent research indicates that Asiatic acid (AA), which is extracted from Trema orientalis L., has potential as a medicinal agent. AA, which was obtained from a methanolic extract of Trema orientalis L. and examined utilizing high-resolution mass spectroscopy (HRMS) analysis, was employed in this investigation. Then, in order to forecast the therapeutic advantages of AA in managing GBM, we conducted an in silico study. Online web servers like SwissADME, pKCSM, and Protox-II were used to assess AA. Then, the major targets of the AA (from Swiss Target Prediction and TargetNet) and GBM (from GeneCards and DisGeNET) were identified. The important genes were then merged into the STRING and ShinyGo databases to examine the protein-protein interaction (PPI) network, gene annotation, and KEGG pathways, with the goal of identifying the core mechanisms involved in GBM management. The top five hub gene targets of the built network (AKT1, SRC, IL-6, TNF, and EGFR) were investigated, along with some contemporaneous additional major targets (PRKCB, GSK3B, ITGB1, BRAF, and PTPN6). These targets were tightly linked to GO activities such as synoviocyte proliferation, cytokine activity, and EGFR tyrosine kinase inhibitor resistance, as well as proteoglycans in cancer-related pathways. Furthermore, a survival study was conducted to assess the chronicity of targets, as well as molecular docking activity between important targets and AA against GBM to determine binding effectiveness. Overall, the study found that AKT1 is the most powerful receptor for AA, having a binding energy of -8.19 kcal/mol, followed by PRKCB (-7.53 kcal/mol). Finally, docking studies suggest that AA has the potential to be an effective treatment for GBM. Furthermore, clinical studies will provide more precise insights into the AA's efficacy as a medicine in the future.
Collapse
Affiliation(s)
- Amit Kumar Singh
- Department of Pharmacognosy, Kunwar Haribansh Singh College of Pharmacy, Jaunpur, Uttar Pradesh 222182, India
| | - Adarsh Kumar Pathak
- Department of Pharmaceutical Chemistry, Ashok Singh Pharmacy College, Jaunpur, Uttar Pradesh 222180, India
| | - Pradeep Kumar
- Department of Botany, MMV, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Anand Kumar Singh
- Department of Chemistry, PG College, Mariahu, VBS Purvanchal University, Jaunpur, Uttar Pradesh 222161, India
| | - Manjeet Kumar Sah Gond
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Rohit Singh Negi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| | - Richa Das
- Department of Biotechnology, Parul Institute of Applied Science, Parul University, Vadodara 391760 Gujarat, India
| | - Shreni Agrawal
- Department of Biotechnology, Parul Institute of Applied Science, Parul University, Vadodara 391760 Gujarat, India
| | - Sunil Kumar Mishra
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India.
| | - Kavindra Nath Tiwari
- Department of Botany, MMV, Banaras Hindu University, Varanasi, Uttar Pradesh 221005, India
| |
Collapse
|
7
|
Zhang P, Wu Y, Chen X, Wang G, Li P, Cao Z, Du H, Zhang C, Du X, Chen G, Zhou Y, Sun Y. TroTNFα, a teleost tumour necrosis factor of golden pompano (Trachinotus ovatus), enhances pathogen clearance and acts as an immune adjuvant. Int J Biol Macromol 2025; 299:140128. [PMID: 39842583 DOI: 10.1016/j.ijbiomac.2025.140128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/07/2025] [Accepted: 01/19/2025] [Indexed: 01/24/2025]
Abstract
Tumour necrosis factor (TNF) is one of the most pivotal factors of the TNF family and plays an essential biological role in immunity. However, the antibacterial function and mechanism of TNFα in teleosts are relatively poorly understood. In this study, a novel TNFα from Trachinotus ovatus (TroTNFα) was characterized. TroTNFα is widely expressed in immune tissues and increased after Vibrio harveyi infection. The recombinant protein TroTNFα facilitated the proliferation and chemotaxis of T. ovatus head kidney lymphocytes, induced apoptosis in human hepatocellular carcinoma cells (HepG2), and enhanced NF-κB promoter activity, whereas mutants with altered conserved receptor binding sites (Phe and Tyr mutated to Ala) lost these functions. Similarly, in vivo research revealed that, compared with the control, TroTNFα overexpression significantly reduced bacterial colonization, whereas the bacterial colonization of the mutants was similar to that of the control. Furthermore, our results showed that TroTNFα increased the vaccine-induced immune responses induced by the DNA vaccine pCTssJ against V. harveyi. Taken together, our results indicate that TroTNFα plays an indispensable role in antibacterial immunity, providing the first evidence that the binding sites (Phe144 and Tyr216) of TroTNFα are crucial in these processes in teleosts and enhances DNA vaccine efficacy as an immune adjuvant.
Collapse
Affiliation(s)
- Panpan Zhang
- Sanya Institute of Breeding and Multiplication, School of Marine Biology and Fisheries, Collaborative Innovation Center of Marine Science and Technology, Hainan University, China
| | - Ying Wu
- Sanya Institute of Breeding and Multiplication, School of Marine Biology and Fisheries, Collaborative Innovation Center of Marine Science and Technology, Hainan University, China
| | - Xiaojuan Chen
- Sanya Institute of Breeding and Multiplication, School of Marine Biology and Fisheries, Collaborative Innovation Center of Marine Science and Technology, Hainan University, China
| | - Guotao Wang
- Sanya Institute of Breeding and Multiplication, School of Marine Biology and Fisheries, Collaborative Innovation Center of Marine Science and Technology, Hainan University, China
| | - Pengfei Li
- Guangxi Key Laboratory of Marine Natural Products and Combinatorial Biosynthesis Chemistry, Guangxi Beibu Gulf Marine Research Center, Guangxi Academy of Sciences, Nanning, China
| | - Zhenjie Cao
- Sanya Institute of Breeding and Multiplication, School of Marine Biology and Fisheries, Collaborative Innovation Center of Marine Science and Technology, Hainan University, China
| | - Hehe Du
- Sanya Institute of Breeding and Multiplication, School of Marine Biology and Fisheries, Collaborative Innovation Center of Marine Science and Technology, Hainan University, China
| | - Chen Zhang
- Sanya Institute of Breeding and Multiplication, School of Marine Biology and Fisheries, Collaborative Innovation Center of Marine Science and Technology, Hainan University, China
| | - Xiangyu Du
- Sanya Institute of Breeding and Multiplication, School of Marine Biology and Fisheries, Collaborative Innovation Center of Marine Science and Technology, Hainan University, China
| | - Guisen Chen
- Sanya Institute of Breeding and Multiplication, School of Marine Biology and Fisheries, Collaborative Innovation Center of Marine Science and Technology, Hainan University, China
| | - Yongcan Zhou
- Sanya Institute of Breeding and Multiplication, School of Marine Biology and Fisheries, Collaborative Innovation Center of Marine Science and Technology, Hainan University, China
| | - Yun Sun
- Sanya Institute of Breeding and Multiplication, School of Marine Biology and Fisheries, Collaborative Innovation Center of Marine Science and Technology, Hainan University, China; Engineering Research Center of Hainan Province for Blue Carbonand Coastal Wetland Conservation and Restoration, China; International Joint Research Center of Hainan Province for Blue Carbon and Coastal Wetland, China.
| |
Collapse
|
8
|
Yang MH, Son NT, Bastos JK, Luyen ND, Linh NN, Ahn KS. Eriodictyol 5-O-methyl ether inhibits prostate cancer progression through targeting STAT3 signaling and inducing apoptosis and paraptosis. Arch Biochem Biophys 2025; 766:110331. [PMID: 39924141 DOI: 10.1016/j.abb.2025.110331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 01/20/2025] [Accepted: 02/06/2025] [Indexed: 02/11/2025]
Abstract
Prostate cancer ranks as one of the most prevalent cancers among men and is a major cause of cancer-related mortality globally This study aims to elucidate the molecular mechanisms underlying the anti-cancer effects of eriodictyol 5-O-methyl ether (ERIO) on prostate cancer cells, focusing on its impact on STAT3 signaling, apoptosis, and paraptosis. ERIO exhibited significant cytotoxicity against DU145, PC-3, and LNCaP cells. It suppressed constitutive and IL-6-induced STAT3 activation by inhibiting the phosphorylation of JAK1, JAK2, and Src kinases. ERIO upregulated SHP-2 expression, leading to the dephosphorylation of STAT3. ERIO induced apoptosis, evidenced by increased caspase-3 and PARP cleavage, and paraptosis, characterized by increased ROS production, decreased mitochondrial membrane potential, and ER stress. The antioxidant NAC reversed the effects of ERIO, highlighting the importance of oxidative stress in its anti-cancer activity. ERIO effectively inhibited prostate cancer cell growth by targeting STAT3 signaling and inducing both apoptosis and paraptosis. These findings suggest that ERIO has significant therapeutic potential for prostate cancer treatment and warrant further investigation in in vivo and clinical studies.
Collapse
Affiliation(s)
- Min Hee Yang
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| | - Ninh The Son
- Institute of Chemistry, Vietnam Academy of Science and Technology (VAST), Hoang Quoc Viet, Caugiay, Hanoi, 10000, Viet Nam; Department of Chemistry, Graduate University of Science and Technology, VAST, Hoang Quoc Viet, Caugiay, Hanoi, 10000, Viet Nam; University of São Paulo (USP), School of Pharmaceutical Sciences of Ribeirão Preto, SP, Brazil.
| | - Jairo Kenupp Bastos
- University of São Paulo (USP), School of Pharmaceutical Sciences of Ribeirão Preto, SP, Brazil.
| | - Nguyen Dinh Luyen
- Institute of Natural Products Chemistry, VAST, Hoang Quoc Viet, Caugiay, Hanoi, 10000, Viet Nam.
| | - Nguyen Ngoc Linh
- Faculty of Pharmacy, Thanh Do University, Kim Chung, Hoai Duc, Hanoi, 10000, Viet Nam.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
9
|
Komleva Y, Shpiliukova K, Bondar N, Salmina A, Khilazheva E, Illarioshkin S, Piradov M. Decoding brain aging trajectory: predictive discrepancies, genetic susceptibilities, and emerging therapeutic strategies. Front Aging Neurosci 2025; 17:1562453. [PMID: 40177249 PMCID: PMC11962000 DOI: 10.3389/fnagi.2025.1562453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
The global extension of human lifespan has intensified the focus on aging, yet its underlying mechanisms remain inadequately understood. The article highlights aspects of genetic susceptibility to impaired brain bioenergetics, trends in age-related gene expression related to neuroinflammation and brain senescence, and the impact of stem cell exhaustion and quiescence on accelerated brain aging. We also review the accumulation of senescent cells, mitochondrial dysfunction, and metabolic disturbances as central pathological processes in aging, emphasizing how these factors contribute to inflammation and disrupt cellular competition defining the aging trajectory. Furthermore, we discuss emerging therapeutic strategies and the future potential of integrating advanced technologies to refine aging assessments. The combination of several methods including genetic analysis, neuroimaging techniques, cognitive tests and digital twins, offer a novel approach by simulating and monitoring individual health and aging trajectories, thereby providing more accurate and personalized insights. Conclusively, the accurate estimation of brain aging trajectories is crucial for understanding and managing aging processes, potentially transforming preventive and therapeutic strategies to improve health outcomes in aging populations.
Collapse
Affiliation(s)
| | | | - Nikolai Bondar
- Research Center of Neurology, Moscow, Russia
- Laboratory of Molecular Virology, First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | - Elena Khilazheva
- Department of Biological Chemistry with Courses in Medical, Research Institute of Molecular Medicine and Pathobiochemistry, Pharmaceutical and Toxicological Chemistry Prof. V.F. Voino-Yasenetsky Krasnoyarsk State Medical University of the Ministry of Healthcare of the Russian Federation, Krasnoyarsk, Russia
| | | | | |
Collapse
|
10
|
Monberg TJ, Pakola SA, Albieri B, Ellebaek E, Donia M, Eefsen RL, Borch TH, Kudling TV, Lorentzen T, Hendel HW, Vestergaard C, Lorentzen C, Holmstroem RB, Arias V, Khammari A, Kistler C, Santos JM, Clubb JHA, Haybout L, Westergaard MCW, Met Ö, Quixabeira DCA, Jirovec E, Havunen R, Sorsa S, Cervera-Carrascon V, Dreno B, Hemminki A, Svane IM. Safety and efficacy of combined treatment with tumor-infiltrating lymphocytes and oncolytic adenovirus TILT-123 in metastatic melanoma. Cell Rep Med 2025; 6:102016. [PMID: 40107242 PMCID: PMC11970381 DOI: 10.1016/j.xcrm.2025.102016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/22/2024] [Accepted: 02/14/2025] [Indexed: 03/22/2025]
Abstract
Tumor-infiltrating lymphocytes (TILs) are effective in the treatment of metastatic melanoma (MM), but toxicity limits its application. TILT-123 (igrelimogene litadenorepvec) is an oncolytic adenovirus producing interleukin-2 (IL-2) and tumor necrosis factor (TNF) upon replication. In this phase 1 trial, 17 patients with metastatic checkpoint inhibitor-resistant melanoma are treated with TILT-123 and TILs without preconditioning chemotherapy or postconditioning IL-2. The treatment is safe and feasible. According to computed tomography (CT), the objective response rate is 11.7% (2/17) and disease control is observed in 35% (6/17), including a partial response lasting >8 months and a durable complete response in a mucosal melanoma patient. According to positron emission tomography (PET), disease control is observed in 7/15 (47%) with minor or partial responses in 4/15 (27%). In the initial TILT-123 monotherapy phase of the trial, disease control is observed in 6/17 (35%) and 10/16 (63%) in CT and PET, respectively. The study demonstrates good tolerability and preliminary efficacy.
Collapse
Affiliation(s)
- Tine J Monberg
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Santeri A Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Progam, University of Helsinki, Helsinki, Finland
| | - Benedetta Albieri
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Eva Ellebaek
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Marco Donia
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Rikke L Eefsen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Troels H Borch
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Tatiana V Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Progam, University of Helsinki, Helsinki, Finland
| | - Torben Lorentzen
- Department of Gastroenterology, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Helle W Hendel
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Cecilie Vestergaard
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Cathrine Lorentzen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Rikke B Holmstroem
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Victor Arias
- Cancer Gene Therapy Group, Translational Immunology Research Progam, University of Helsinki, Helsinki, Finland
| | - Amir Khammari
- Nantes Université, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302/EMR6001, 44000 Nantes, France
| | | | - João M Santos
- Cancer Gene Therapy Group, Translational Immunology Research Progam, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland
| | - James H A Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Progam, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Lyna Haybout
- Cancer Gene Therapy Group, Translational Immunology Research Progam, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Marie C W Westergaard
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730 Herlev, Denmark
| | - Özcan Met
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730 Herlev, Denmark; Department of Health Technology, Technical University of Denmark, 2800 Lyngby, Denmark
| | - Dafne C A Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Progam, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Elise Jirovec
- Cancer Gene Therapy Group, Translational Immunology Research Progam, University of Helsinki, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Progam, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Suvi Sorsa
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Progam, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Brigitte Dreno
- Nantes Université, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302/EMR6001, 44000 Nantes, France
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Progam, University of Helsinki, Helsinki, Finland; TILT Biotherapeutics Ltd, Helsinki, Finland; Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730 Herlev, Denmark.
| |
Collapse
|
11
|
Ebrahimi F, Gholizadeh M, Sahebalam H. Genome-wide study for signatures of selection identifies genomic regions and candidate genes associated with milk traits in sheep. Mamm Genome 2025; 36:140-150. [PMID: 39904907 DOI: 10.1007/s00335-025-10107-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 01/24/2025] [Indexed: 02/06/2025]
Abstract
Milk production traits in sheep are influenced by complex genetic factors, and understanding these traits requires the identification of candidate genes under selection. This study employed two methods, FST and XP-EHH, to identify selection signatures and candidate genes associated with milk production traits in sheep. For this purpose, 9 different breeds from the Sheep HapMap dataset generated by the International Sheep Genomics Consortium (ISGC) based on analysis of the Ovine SNP50 BeadChip were used. The dairy breeds included Brown East Friesian (n = 39), Milk Lacaune (n = 103), Chios (n = 23), Churra (n = 120), and Comisana (n = 24), while the non-dairy breeds included Afshari (n = 37), Moghani (n = 34), Galway (n = 49), and Australian Suffolk (n = 109). Genomic regions in the top 0.1 percentile of FST values revealed 71 genes, while regions with the highest positive XP-EHH values identified 69 genes. Five overlapping genes-DHRS3, TNFRSF1B, AADACL4, ARHGEF11, and LRRC71-were detected by both methods, highlighting their relevance to milk production. Several candidate genes in regions identified from FST, such as PER2, SH3PXD2A, TMEM117, DDX6, PDCD11, CALHM2, and CALHM3, have been previously associated with milk production traits. Notably, CRABP2, PEAR1, PGM1, ALG6, COX15, and OAT were identified in regions with high XP-EHH values in the dairy group. Gene ontology analysis indicated that the identified genes are enriched in pathways related to chemokine receptor activity, gap junction channel activity, and gap junction-mediated intercellular transport, as well as cellular components like the connexin complex. Further studies on these genes may improve understanding of the genetic architecture of milk production traits in sheep.
Collapse
Affiliation(s)
- Fatemeh Ebrahimi
- Department of Animal Science, Faculty of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Resources University, Sari, Iran
| | - Mohsen Gholizadeh
- Department of Animal Science, Faculty of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Resources University, Sari, Iran.
| | - Hamid Sahebalam
- Department of Animal Science, Faculty of Animal Science and Fisheries, Sari Agricultural Sciences and Natural Resources University, Sari, Iran
| |
Collapse
|
12
|
Rodponthukwaji K, Khowawisetsut L, Limjunyawong N, Kunwong N, Duangchan K, Sripinitchai S, Sathornsumetee S, Nguyen T, Srisawat C, Punnakitikashem P. Enhanced Anticancer Effects Through Combined Therapeutic Model of Macrophage Polarization and Cancer Cell Apoptosis by Multifunctional Lipid Nanocomposites. J Biomed Mater Res A 2025; 113:e37886. [PMID: 39972623 DOI: 10.1002/jbm.a.37886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 01/20/2025] [Accepted: 02/07/2025] [Indexed: 02/21/2025]
Abstract
Although the mono-anticancer therapy approach particularly directly targeting tumors is still common, this conventional method is generally deemed not effective and insufficient. In tumor microenvironment (TME), tumor-associated macrophages (TAMs, referred to as M2-polarized) play a crucial role in creating an immunosuppressive TME, contributing to various pro-tumorigenic effects. A promising strategy to inhibit tumor growth involves re-educating M2 macrophages into tumoricidal macrophages (M1). Therefore, combining macrophage reprogramming with cancer cell death induction in a single modality may offer synergistic benefits in cancer therapy. Here, we engineered a lipid-based delivery platform capable of co-delivering resiquimod (R848) and polyinosinic: polycytidylic acid (PIC). R848 in our nanosystem effectively triggered M2-to-M1 repolarization, as evidenced by the upregulation of M1 marker genes (TNF, IL6), the release of proinflammatory cytokines (TNF-α and IL-6), and the downregulation of the M2 marker gene, MRC1. On the other hand, the presence of PIC increased caspase-3/7 activity leading to cancer cell death through the apoptotic pathway. This nanocarrier system established a multifunctional platform to enhance the anticancer effect. The synergistic effect of repolarized macrophages in combination with the induction of apoptosis, facilitated by our nanomedicine, was evident in a co-culture system of macrophage and cancer cells, showing a significant increase in cancer cell death compared to individual treatments. These findings attractively demonstrated the potential of our multifunctional lipid nanoparticles as therapeutic agents for anticancer treatment by modulating the tumor immune microenvironment and simultaneously increasing cancer cell cytotoxicity.
Collapse
Affiliation(s)
- Kamonlatth Rodponthukwaji
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ladawan Khowawisetsut
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence for Microparticle and Exosome in Diseases, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nathachit Limjunyawong
- Siriraj Center of Research Excellence in Allergy and Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Natsuda Kunwong
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kongpop Duangchan
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sirinapa Sripinitchai
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sith Sathornsumetee
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tam Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Chatchawan Srisawat
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Primana Punnakitikashem
- Department of Biochemistry, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Center of Research Excellence in Theranostic Nanomedicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| |
Collapse
|
13
|
Szóstak N, Budnik M, Tomela K, Handschuh L, Samelak-Czajka A, Pietrzak B, Schmidt M, Kaczmarek M, Galus Ł, Mackiewicz J, Mackiewicz A, Kozlowski P, Philips A. Exploring correlations between gut mycobiome and lymphocytes in melanoma patients undergoing anti-PD-1 therapy. Cancer Immunol Immunother 2025; 74:110. [PMID: 39998665 PMCID: PMC11861499 DOI: 10.1007/s00262-024-03918-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 12/02/2024] [Indexed: 02/27/2025]
Abstract
Research has shown that the microbiome can influence how the immune system responds to melanoma cells, affecting the course of the disease and the outcome of the therapy. Here, we used the metagenomic approach and flow cytometry analyses of blood cells to discover correlations between gut fungi of metastatic melanoma patients enrolled in anti-PD-1 therapy and lymphocytes in their blood.We analyzed the patterns of associations before the first administration of anti-PD-1 therapy (BT, n = 61) and in the third month of the therapy (T3, n = 37), allowing us to track changes during treatment. To understand the possible impact of gut fungi on the efficacy of anti-PD-1 therapy, we analyzed the associations in clinical beneficiaries (CB, n = 37) and non-beneficiaries (NB, n = 24), as well as responders (R, n = 28) and non-responders (NR, n = 33).Patients with LDH < 338 units/L, overall survival (OS) > 12, CB, as well as R, had lower levels of Shannon diversity (p = 0.02, p = 0.05, p = 0.05, and p = 0.03, respectively). We found that the correlation pattern between intestinal fungi and lymphocytes was specific to the type of response, positive or negative. When comparing CB and NB groups, correlations with opposite directions were detected for C. albicans, suggesting a response-specific immune reaction. For CB, M. restricta exhibited a set of correlations with different types of lymphocytes, with prevalent positive correlations, suggesting a robust immune response in the CB group. This result extends our former research, where M. restricta and C. albicans were associated with an increased risk of melanoma progression and a poorer response to anti-PD-1 treatment.
Collapse
Affiliation(s)
- Natalia Szóstak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland.
| | - Michał Budnik
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Katarzyna Tomela
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866, Poznan, Poland
| | - Luiza Handschuh
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Anna Samelak-Czajka
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Bernadeta Pietrzak
- Department of Food Biotechnology and Microbiology, Poznan University of Life Sciences, Poznan, Poland
| | - Marcin Schmidt
- Department of Food Biotechnology and Microbiology, Poznan University of Life Sciences, Poznan, Poland
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866, Poznan, Poland
| | - Łukasz Galus
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866, Poznan, Poland
- Department of Medical and Experimental Oncology, Institute of Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Jacek Mackiewicz
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866, Poznan, Poland
- Department of Medical and Experimental Oncology, Institute of Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Andrzej Mackiewicz
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866, Poznan, Poland
| | - Piotr Kozlowski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Anna Philips
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
14
|
Zhang C, Chen Z, Qi G, Tian Y, Zheng X, Diao X, Kong J, Ju X, Li J, Dong S, Jin Y. Smart and Noninvasive SERS Immunosensors for Monitoring Dynamic Expression of Cytokines during Cell Pyroptosis. Anal Chem 2025; 97:1783-1791. [PMID: 39810416 DOI: 10.1021/acs.analchem.4c05539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Accompanying the occurrence of inflammatory reaction to release cytokines, pyroptosis can activate an immune response for resistance against cancer. Consequently, elevated levels of cytokines released by cancer cells are highly correlated with the effectiveness of cancer treatment. Herein, a noninvasive surface-enhanced Raman spectroscopy (SERS) immunosensor was developed to sensitively and specifically measure the tumor necrosis factor-α (TNF-α), a proinflammatory cytokine, during the cell pyroptosis process. The sandwiched structure of the sensor is functionalized with a TNF-α binding antibody for detecting TNF-α at concentrations as low as 1 pg/mL. Importantly, electrical stimulation (ES) can fleetly trigger cancer cell pyroptosis to induce the overexpression of receptor interacting protein 3 (RIP3), which is a significant protein that regulates the inflammatory response. The overexpression of RIP3 can activate caspase-1 to promote the upregulation of cytokine levels. Notably, the cytokine levels of TNF-α released from cancer cells (MCF-7 cells) were apparently higher than those of normal cells (MCF-10A cells) during pyroptosis detected by the SERS immunosensors. Due to its obvious superiorities of simple fabrication and fast readout without sample pretreatment, the developed SERS platform has a potential application value for diagnosis and treatment of cancer.
Collapse
Affiliation(s)
- Chenyu Zhang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China
| | - Zutao Chen
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, PR China
| | - Guohua Qi
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, PR China
| | - Yu Tian
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Xiongjian Zheng
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, PR China
| | - Xingkang Diao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China
| | - Jiao Kong
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China
| | - Xingkai Ju
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China
| | - Jing Li
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China
| | - Shaojun Dong
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China
| | - Yongdong Jin
- Guangdong Key Laboratory of Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, PR China
| |
Collapse
|
15
|
Sutera S, Furchì OA, Pentenero M. Investigating Tumor-Infiltrating Lymphocytes in the Microenvironment of Oral Squamous Cell Carcinoma (OSCC) and Oral Potentially Malignant Disorders (OPMDs): Can They Shift Our Perspective? A Scoping Review. J Clin Med 2025; 14:606. [PMID: 39860614 PMCID: PMC11766165 DOI: 10.3390/jcm14020606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/17/2024] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Tumor-infiltrating lymphocytes (TILs) play a crucial role in the tumor microenvironment (TME), influencing the progression, prognosis, and response to treatment in oral squamous cell carcinoma (OSCC) and its precursors, oral potentially malignant disorders (OPMDs). This scoping review assesses the current literature on TILs in the TME of OSCC and OPMDs, aiming to identify trends and gaps in the research. Methods: A comprehensive search was performed in PubMed, using the following query terms: "Tumor Microenvironment AND (mouth neoplasms OR oral lichen OR leukoplakia OR oral lichenoid OR dysplasia OR GVHD OR lupus)". Based on the inclusion criteria, we selected in vivo human original research and clinical observational studies that focused on TILs within the TME of OSCC and OPMDs. Results: Out of 1152 results in PubMed, 58 studies were selected and analyzed. These studies investigated various TILs, including T cells, B cells, and natural killer (NK) cells. Of these, 47 studies focused on the OSCC TME, 4 examined the OPMDs ME, and 7 compared OSCC TME and OPMDs ME. Discussion: While TILs in OSCC have been extensively studied, research on infiltrating lymphocytes in OPMDs is still limited. In OSCC, CD8+ T cells, T helper 1 cells, and NK cells are associated with strong antitumor activity, whereas CD4+ T cells, including T helper 2 and regulatory T cells, are linked to protumoral effects. B cells remain less explored due to their low frequency in the TME. In OPMDs, trends suggest an increase in activated CD8+ T cells in OLP and lower NK cell numbers compared to OSCC, which may contribute to malignant transformation. Understanding the spatial distribution and activation status of TILs within the TME is essential for deciphering their role. The variability in TIL composition highlights the complexity of the TME. Conclusions: Current knowledge remains preliminary, though it highlights the crucial role of TILs in carcinogenesis and OSCC. A more in-depth understanding could improve diagnostic and therapeutic strategies, including the assessment of the risk of malignant transformation in OPMDs.
Collapse
Affiliation(s)
- Samuele Sutera
- Oral Medicine and Oral Oncology Unit, Department of Oncology, University of Turin, 10043 Turin, Italy; (O.A.F.); (M.P.)
| | | | | |
Collapse
|
16
|
Tahrali I, Yucel E, Turkkan E, Aycicek A, Unuvar A, Cinar S, Deniz G. The effects of bone marrow humoral components of B-cell acute lymphoblastic leukemia patients on natural killer cell suppression. Immunol Res 2025; 73:31. [PMID: 39808387 DOI: 10.1007/s12026-024-09577-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/18/2024] [Indexed: 01/16/2025]
Abstract
B-cell acute lymphoblastic leukemia (B-ALL) is the most common form of cancer diagnosed in children. While the majority of patients survive with conventional treatment, chemotherapeutic agents have adverse effects and the potential for relapse persists even after full recovery. Given their pivotal function in anti-cancer immunity, there has been a surge in research exploring the potential of natural killer (NK) cells in immunotherapy, which has emerged as a promising avenue for treating leukemia. Nevertheless, the efficacy of NK cell immunotherapy is less pronounced than expected, which suggests the external conditions that affect NK cell functions after the administration to patients with leukemia. In this study, the effects of humoral components in the bone marrow humoral components of B-ALL patients on healthy NK cells were investigated. Healthy peripheral blood mononuclear cells were cultured with and without bone marrow-derived plasma samples of B-ALL patients. The expression of PD-1 and IL-10 were found to be increased whereas the proliferative capacities of NK cells were found to be decreased at the presence of B-ALL plasma samples. Moreover, high IL-10 versus low IL-18 levels were detected in bone marrow plasma samples of B-ALL patients. These findings indicate that humoral components in the bone marrow of B-ALL patients exert a suppressive effect on NK cell functions.
Collapse
Affiliation(s)
- Ilhan Tahrali
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Türkiye
- Department of Pharmaceutical Microbiology, School of Pharmacy, Istanbul Medipol University, Istanbul, Türkiye
| | - Esra Yucel
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Türkiye
| | - Emine Turkkan
- Istanbul Okmeydani Health Application and Research Center, Pediatric Hematology and Oncology, University of Health Sciences, Istanbul, Türkiye
| | - Ali Aycicek
- Department of Pediatric Hematology and Oncology, Çam and Sakura City Hospital, Istanbul, Türkiye
| | - Aysegul Unuvar
- Division of Pediatric Hematology and Oncology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Türkiye
| | - Suzan Cinar
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Türkiye
| | - Gunnur Deniz
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Türkiye.
| |
Collapse
|
17
|
Vijayanathan Y, Ho IAW. The Impact of Metabolic Rewiring in Glioblastoma: The Immune Landscape and Therapeutic Strategies. Int J Mol Sci 2025; 26:669. [PMID: 39859381 PMCID: PMC11765942 DOI: 10.3390/ijms26020669] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/06/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Glioblastoma (GBM) is an aggressive brain tumor characterized by extensive metabolic reprogramming that drives tumor growth and therapeutic resistance. Key metabolic pathways, including glycolysis, lactate production, and lipid metabolism, are upregulated to sustain tumor survival in the hypoxic and nutrient-deprived tumor microenvironment (TME), while glutamine and tryptophan metabolism further contribute to the aggressive phenotype of GBM. These metabolic alterations impair immune cell function, leading to exhaustion and stress in CD8+ and CD4+ T cells while favoring immunosuppressive populations such as regulatory T cells (Tregs) and M2-like macrophages. Recent studies emphasize the role of slow-cycling GBM cells (SCCs), lipid-laden macrophages, and tumor-associated astrocytes (TAAs) in reshaping GBM's metabolic landscape and reinforcing immune evasion. Genetic mutations, including Isocitrate Dehydrogenase (IDH) mutations, Epidermal Growth Factor Receptor (EGFR) amplification, and Phosphotase and Tensin Homolog (PTEN) loss, further drive metabolic reprogramming and offer potential targets for therapy. Understanding the relationship between GBM metabolism and immune suppression is critical for overcoming therapeutic resistance. This review focuses on the role of metabolic rewiring in GBM, its impact on the immune microenvironment, and the potential of combining metabolic targeting with immunotherapy to improve clinical outcomes for GBM patients.
Collapse
Affiliation(s)
- Yuganthini Vijayanathan
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore 308433, Singapore;
| | - Ivy A. W. Ho
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore 308433, Singapore;
- Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Physiology, National University of Singapore, Singapore 117593, Singapore
| |
Collapse
|
18
|
Haysom‐McDowell A, Paudel KR, Yeung S, Kokkinis S, El Sherkawi T, Chellappan DK, Adams J, Dua K, De Rubis G. Recent trends and therapeutic potential of phytoceutical-based nanoparticle delivery systems in mitigating non-small cell lung cancer. Mol Oncol 2025; 19:15-36. [PMID: 39592417 PMCID: PMC11705733 DOI: 10.1002/1878-0261.13764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/20/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
Lung cancer is the leading cause of cancer death globally, with non-small cell lung cancer accounting for the majority (85%) of cases. Standard treatments including chemotherapy and radiotherapy present multiple adverse effects. Medicinal plants, used for centuries, are traditionally processed by methods such as boiling and oral ingestion, However, water solubility, absorption, and hepatic metabolism reduce phytoceutical bioavailability. More recently, isolated molecular compounds from these plants can be extracted with these phytoceuticals administered either individually or as an adjunct with standard therapy. Phytoceuticals have been shown to alleviate symptoms, may reduce dosage of chemotherapy and, in some cases, enhance pharmaceutical mechanisms. Research has identified many phytoceuticals' actions on cancer-associated pathways, such as oncogenesis, the tumour microenvironment, tumour cell proliferation, metastasis, and apoptosis. The development of novel nanoparticle delivery systems such as solid lipid nanoparticles, liquid crystalline nanoparticles, and liposomes has enhanced the bioavailability and targeted delivery of pharmaceuticals and phytoceuticals. This review explores the biological pathways associated with non-small cell lung cancer, a diverse range of phytoceuticals, the cancer pathways they act upon, and the pros and cons of several nanoparticle delivery systems.
Collapse
Affiliation(s)
- Adam Haysom‐McDowell
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoAustralia
- Australian Research Consortium in Complementary and Integrative Medicine, School of Public HealthUniversity of Technology SydneyUltimoAustralia
| | - Keshav Raj Paudel
- Australian Research Consortium in Complementary and Integrative Medicine, School of Public HealthUniversity of Technology SydneyUltimoAustralia
- Centre for Inflammation Centenary Institute, Faculty of Science, School of Life SciencesUniversity of Technology SydneyAustralia
| | - Stewart Yeung
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoAustralia
- Australian Research Consortium in Complementary and Integrative Medicine, School of Public HealthUniversity of Technology SydneyUltimoAustralia
| | - Sofia Kokkinis
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoAustralia
- Australian Research Consortium in Complementary and Integrative Medicine, School of Public HealthUniversity of Technology SydneyUltimoAustralia
| | - Tammam El Sherkawi
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoAustralia
- Australian Research Consortium in Complementary and Integrative Medicine, School of Public HealthUniversity of Technology SydneyUltimoAustralia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of PharmacyInternational Medical UniversityKuala LumpurMalaysia
| | - Jon Adams
- Australian Research Consortium in Complementary and Integrative Medicine, School of Public HealthUniversity of Technology SydneyUltimoAustralia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoAustralia
- Australian Research Consortium in Complementary and Integrative Medicine, School of Public HealthUniversity of Technology SydneyUltimoAustralia
| | - Gabriele De Rubis
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoAustralia
- Australian Research Consortium in Complementary and Integrative Medicine, School of Public HealthUniversity of Technology SydneyUltimoAustralia
| |
Collapse
|
19
|
Akyigit A, Arslan Solmaz O, Kalayci M, Sakallioglu O, Duzer S, Ozercan İH, Keles E, Karlidag T, Kaygusuz I, Yalcin S. The role of irisin and cytokines in the etiology of parotid tumors. Biotech Histochem 2025; 100:32-39. [PMID: 39772907 DOI: 10.1080/10520295.2025.2450406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
This study explores the role of irisin and interleukins in parotid tumors by determining the tissue staining intensity of irisin, the salivary and plasma levels of irisin, and the plasma levels of IL-4, IL-6, IL-10 and TNF-alpha in individuals with parotid tumors. Forty-eight patients and forty healthy individuals were included to the study and allocated into four group. Benign Group I (pleomorphic adenoma), Group II (Warthin's tumor), Group III (mucoepidermoid carcinoma) and Group IV (benign parotid control group, healthy control group). Parotid tissue, plasma and saliva samples were collected from each of the patients with parotid tumors, while plasma and saliva samples were collected from the healthy individuals. Normal parotid tissue for histologic evaluation was obtained from unaffected areas in patients with parotid tumors. The levels of irisin in the plasma and saliva were significantly lower in the parotid tumors, while the plasma levels of IL-6 and TNF-alpha were higher in patients with parotid tumors, but no statistically significant difference in IL-4 and IL-10 levels was found. The histopathological intensity of FNDC-5/irisin staining was significantly decreased in the parotid tumor tissues when compared to the benign parotid control group with normal parotid tissue. The low histopathological tissue staining intensity and plasma and salivary levels of irisin suggest that irisin may be a protective protein in parotid tumors.
Collapse
Affiliation(s)
- Abdulvahap Akyigit
- Department of Ear Nose and Throat, Firat University Faculty of Medicine, Elazig, Turkey
| | - Ozgen Arslan Solmaz
- Clinic of Medical Pathology, Elazig Fethi Sekin City Hospital, Elazig, Turkey
| | - Mehmet Kalayci
- Clinic of Medical Biochemistry, Duzce Ataturk State Hospital, Duzce, Turkey
| | - Oner Sakallioglu
- Clinic of Ear Nose and Throat, Elazig Fethi Sekin City Hospital, Elazig, Turkey
| | - Sertac Duzer
- Clinic of Ear Nose and Throat, Elazig Fethi Sekin City Hospital, Elazig, Turkey
| | | | - Erol Keles
- Department of Ear Nose and Throat, Firat University Faculty of Medicine, Elazig, Turkey
| | - Turgut Karlidag
- Department of Ear Nose and Throat, Firat University Faculty of Medicine, Elazig, Turkey
| | - Irfan Kaygusuz
- Department of Ear Nose and Throat, Firat University Faculty of Medicine, Elazig, Turkey
| | - Sinasi Yalcin
- Department of Ear Nose and Throat, Firat University Faculty of Medicine, Elazig, Turkey
| |
Collapse
|
20
|
Tyagi S, Tyagi N, Singh A, Gautam A, Singh A, Jindal S, Singh RP, Chaturvedi R, Kushwaha HR. Linking COVID-19 and cancer: Underlying mechanism. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167563. [PMID: 39510388 DOI: 10.1016/j.bbadis.2024.167563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/13/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), lead to a global health crisis with a spectrum of clinical manifestations. A potentially vulnerable category for SARS-CoV-2 infection was identified in patients with other medical conditions. Intriguingly, parallels exist between COVID-19 and cancer at the pathophysiological level, suggesting a possible connection between them. This review discusses all possible associations between COVID-19 and cancer. Expression of receptors like angiotensin-converting enzyme 2 (ACE2) and transmembrane protease serine 2 (TMPRSS2) increases COVID-19 susceptibility. SARS-CoV-2 infection might increase cancer susceptibility and accelerate cancer progression through mechanisms involving cytokine storm, tissue hypoxia, impaired T-cell responses, autophagy, neutrophil activation, and oxidative stress. These mechanisms collectively contribute to immune suppression, hindered apoptosis, and altered cellular signaling in the tumor microenvironment, creating conditions favorable for tumor growth, metastasis, and recurrence. Approved vaccines and their impact on cancer patients along-with new clinical trials are also described.
Collapse
Affiliation(s)
- Sourabh Tyagi
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Nipanshi Tyagi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Anu Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Akanksha Gautam
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Awantika Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India
| | - Shelja Jindal
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India
| | - Rana P Singh
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India; School of Life Sciences, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Rupesh Chaturvedi
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India; School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| | - Hemant Ritturaj Kushwaha
- Special Centre for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India; School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
21
|
Kappen J, Abdel-Rahman O. Advances in pharmacotherapy for the treatment of peritoneal metastases from colorectal cancer. Expert Opin Pharmacother 2025; 26:17-30. [PMID: 39604139 DOI: 10.1080/14656566.2024.2435946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/26/2024] [Indexed: 11/29/2024]
Abstract
INTRODUCTION Patients with peritoneal metastasis (PM) from colorectal cancer (CRC) typically have a poor prognosis with historically few treatment options. Cytoreductive surgery (CRS) is the mainstay of treatment to remove macrometastases into the peritoneum, but residual micrometastases are often left behind. Systemic chemotherapy remains a cornerstone of treatment for micrometastases, but intraperitoneal therapy offers advantages including higher local dose concentration with fewer systemic side effects from treatment. AREAS COVERED This review covers advancements in the routes and types of pharmacotherapies for PM in CRC. EXPERT OPINION More evidence is needed to justify HIPEC with CRS as the standard of care treatment modality for patients with resectable PM in CRC. New therapies such as oncolytic viruses, biologics, and small-molecule inhibitors may become additional treatment modalities for PM.
Collapse
Affiliation(s)
- Janson Kappen
- Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Omar Abdel-Rahman
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| |
Collapse
|
22
|
Calaf GM, Roy D, Jara L, Romero C, Crispin LA. Genes Associated with the Immune System Affected by Ionizing Radiation and Estrogen in an Experimental Breast Cancer Model. BIOLOGY 2024; 13:1078. [PMID: 39765744 PMCID: PMC11673214 DOI: 10.3390/biology13121078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
Breast cancer is a global health issue that, when in the metastasis stage, is characterized by the lack of estrogen receptor-α, the progesterone receptor, and human epidermal growth receptor expressions. The present study analyzed the differential gene expression related to the immune system affected by ionizing radiation and estrogen in cell lines derived from an experimental breast cancer model that was previously developed; where the immortalized human breast epithelial cell line MCF-10F, a triple-negative breast cancer cell line, was exposed to low doses of high linear energy transfer α particle radiation (150 keV/μm), it subsequently grew in the presence or absence of 17β-estradiol. Results indicated that interferon-related developmental regulator 1 gene expression was affected in the estrogen-treated cell line; this interferon, as well as the Interferon-Induced Transmembrane protein 2, and the TNF alpha-induced Protein 6 gene expression levels were higher than the control in the Alpha3 cell line. Furthermore, the interferon-related developmental regulator 1, the Interferon-Induced Transmembrane protein 2, the TNF alpha-induced Protein 6, the Nuclear Factor Interleukin 3-regulated, and the Interferon-Gamma Receptor 1 showed high expression levels in the Alpha5 cell line, and the Interferon Regulatory Factor 6 was high in the Tumor2 cell line. Additionally, to further strengthen these data, publicly available datasets were analyzed. This analysis was conducted to assess the correlation between estrogen receptor alpha expression and the genes mentioned above in breast cancer patients, the differential gene expression between tumor and normal tissues, the immune infiltration level, the ER status, and the survival outcome adjusted by the clinical stage factor. It can be concluded that the genes of the interferon family and Tumor Necrosis factors can be potential therapeutic targets for breast cancer, since they are active before tumor formation as a defense of the body under radiation or estrogen effects.
Collapse
Affiliation(s)
- Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile;
| | - Debasish Roy
- Department of Natural Sciences, Hostos College of the City University of New York, Bronx, NY 10451, USA;
| | - Lilian Jara
- Laboratorio de Genética Humana, Programa de Genética Humana, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile;
| | - Carmen Romero
- Laboratorio de Endocrinología, Facultad de Medicina, Universidad de Chile, Santiago 8380000, Chile;
| | - Leodan A. Crispin
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile;
| |
Collapse
|
23
|
Xue B, Qualls C, Lanthiez A, Lu QY, Yang J, Lee RP, Neis P, Mao JT. The Effects of a Grape Seed Procyanidin Extract on Cytochrome P450 3A4 Activity and Inflammatory Mediators in the Lungs of Heavy Active and Former Smokers. Int J Mol Sci 2024; 25:13105. [PMID: 39684816 DOI: 10.3390/ijms252313105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Grape seed procyanidin extract (GSE) is widely used to promote cardiovascular health and has purported anti-inflammatory properties. Chronic inflammation in the lungs caused by environmental toxins such as tobacco smoking plays a pivotal role in lung cancer development. In a modified phase I lung cancer chemoprevention study conducted in heavy active and former smokers using leucoselect phytosome (LP), a standardized grape seed procyanidin extract complexed with soy phospholipids to enhance bioavailability, three months of LP treatment favorably modulated a variety of surrogate endpoint biomarkers, including markers of cell proliferation. In this correlative study, we further analyzed the effects of LP on cytochrome P450 3A4 (CYP3A4) activities by comparing the endogenous conversions of cortisol and cortisone to 6-beta-hydroxycortisol and 6-beta-hydroxycortisone, respectively, before and after LP treatment and the anti-inflammatory effects of LP in the lung microenvironment of these participants by comparing a profile of inflammatory cytokines and chemokines in matched pre- and post-treatment bronchoalveolar lavage (BAL) fluids. LP treatment did not significantly alter CYP3A4 activity, and three months of LP treatment significantly decreased tumor necrosis factor (TNF), C-C Motif Chemokine Ligand 3 (CCL3) and granzyme B in BAL fluids. Furthermore, post-LP-treatment BAL fluids significantly reduced migration/invasion of various human lung neoplastic cells in vitro. Our findings support the anti-inflammatory effects of GSE/LP in the lung microenvironment and its potential utility for reducing cancerizing forces, as well as driving forces for other common respiratory diseases such as chronic obstructive pulmonary disease and asthma, in the lungs of heavy former and active smokers.
Collapse
Affiliation(s)
- Bingye Xue
- Pulmonary, Critical Care and Sleep Section, New Mexico Veterans Administration Health Care System, University of New Mexico, Albuquerque, NM 87108, USA
| | - Clifford Qualls
- Biostatistics, Biomedical Research Institute of New Mexico, New Mexico Veterans Administration Health Care System, University of New Mexico, Albuquerque, NM 87108, USA
| | - Alexander Lanthiez
- Pulmonary and Critical Care Section, Veterans Administration San Diego Healthcare System, University of California San Diego, La Jolla, CA 92161, USA
| | - Qing-Yi Lu
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Jieping Yang
- UCLA Center for Human Nutrition, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Ru-Po Lee
- UCLA Center for Human Nutrition, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Patricia Neis
- Pulmonary, Critical Care and Sleep Section, New Mexico Veterans Administration Health Care System, University of New Mexico, Albuquerque, NM 87108, USA
| | - Jenny T Mao
- Pulmonary, Critical Care and Sleep Section, New Mexico Veterans Administration Health Care System, University of New Mexico, Albuquerque, NM 87108, USA
- Pulmonary and Critical Care Section, Veterans Administration San Diego Healthcare System, University of California San Diego, La Jolla, CA 92161, USA
| |
Collapse
|
24
|
Paterson C, Vargis E. Applying low levels of strain to model nascent phenomenon of retinal pathologies. LAB ON A CHIP 2024; 24:5338-5346. [PMID: 39575534 PMCID: PMC11884429 DOI: 10.1039/d4lc00205a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/08/2025]
Abstract
Age-related macular degeneration (AMD) is a leading cause of vision loss in aging populations. A better understanding of the mechanisms of the disease, especially at early stages, could elucidate new treatment targets. One characteristic of AMD is strain on the retinal pigment epithelium (RPE), a crucial layer of the retina. This strain can be caused by physical phenomena like waste aggregation underneath the RPE, drusen formation, or leaky blood vessels that infiltrate the retina during choroidal neovascularization (CNV). It is not well understood how strain affects RPE cell function. Most models generate equibiaxial strain or higher levels of strain that are not representative of early stages of AMD. To overcome these issues, we engineered a device to cause controlled, low amounts of localized, radial strain (maximum ∼1.4%). This strain level is more mimetic to what occurs during aging or at the beginning of physical disruptions experienced during AMD. To evaluate how RPE cells respond to this physical stimulus, primary porcine RPE cells were exposed to low levels of strain applied by our custom-made device. Cell secretions and genetic expression were analyzed to determine how proteins linked to drusen and CNV are affected. The results indicate that this low amount of strain does not immediately initiate angiogenesis but causes changes in mRNA expression of amyloid precursor protein (APP), which plays a role in retinal health and drusen accumulation. This research offers insight into AMD progression as well as the health of other organs, including the brain.
Collapse
Affiliation(s)
- Chase Paterson
- Biological Engineering, Utah State University College of Engineering, 4105 Old Main Hill, ENGR 402, Logan, Utah, USA.
| | - Elizabeth Vargis
- Biological Engineering, Utah State University College of Engineering, 4105 Old Main Hill, ENGR 402, Logan, Utah, USA.
| |
Collapse
|
25
|
Esteban-Medina M, de la Oliva Roque VM, Herráiz-Gil S, Peña-Chilet M, Dopazo J, Loucera C. drexml: A command line tool and Python package for drug repurposing. Comput Struct Biotechnol J 2024; 23:1129-1143. [PMID: 38510973 PMCID: PMC10950807 DOI: 10.1016/j.csbj.2024.02.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/22/2024] Open
Abstract
We introduce drexml, a command line tool and Python package for rational data-driven drug repurposing. The package employs machine learning and mechanistic signal transduction modeling to identify drug targets capable of regulating a particular disease. In addition, it employs explainability tools to contextualize potential drug targets within the functional landscape of the disease. The methodology is validated in Fanconi Anemia and Familial Melanoma, two distinct rare diseases where there is a pressing need for solutions. In the Fanconi Anemia case, the model successfully predicts previously validated repurposed drugs, while in the Familial Melanoma case, it identifies a promising set of drugs for further investigation.
Collapse
Affiliation(s)
- Marina Esteban-Medina
- Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, Seville, Spain
| | - Víctor Manuel de la Oliva Roque
- Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, Seville, Spain
| | - Sara Herráiz-Gil
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), U714, Madrid, Spain
- Departamento de Bioingeniería, Universidad Carlos III de Madrid (UC3M), Madrid, Spain
- Regenerative Medicine and Tissue Engineering Group, Instituto de Investigación Sanitaria-Fundación Jiménez Díaz University Hospital (IIS-FJD), Madrid, Spain
- Epithelial Biomedicine Division, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain
| | - María Peña-Chilet
- Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
- Platform of Big Data, AI and Biostatistics, Health Research Institute La Fe (IISLAFE), Valencia, Spain
| | - Joaquín Dopazo
- Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), U715, Seville, Spain
- FPS/ELIXIR-es, Hospital Virgen del Rocío, Seville, Spain
| | - Carlos Loucera
- Platform for Computational Medicine, Andalusian Public Foundation Progress and Health-FPS, Seville, Spain
- Computational Systems Medicine, Institute of Biomedicine of Seville (IBIS), Hospital Virgen del Rocío, Seville, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII), U715, Seville, Spain
| |
Collapse
|
26
|
Ababneh O, Nishizaki D, Kato S, Kurzrock R. Tumor necrosis factor superfamily signaling: life and death in cancer. Cancer Metastasis Rev 2024; 43:1137-1163. [PMID: 39363128 PMCID: PMC11554763 DOI: 10.1007/s10555-024-10206-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/13/2024] [Indexed: 10/05/2024]
Abstract
Immune checkpoint inhibitors have shaped the landscape of cancer treatment. However, many patients either do not respond or suffer from later progression. Numerous proteins can control immune system activity, including multiple tumor necrosis factor (TNF) superfamily (TNFSF) and TNF receptor superfamily (TNFRSF) members; these proteins play a complex role in regulating cell survival and death, cellular differentiation, and immune system activity. Notably, TNFSF/TNFRSF molecules may display either pro-tumoral or anti-tumoral activity, or even both, depending on tumor type. Therefore, TNF is a prototype of an enigmatic two-faced mediator in oncogenesis. To date, multiple anti-TNF agents have been approved and/or included in guidelines for treating autoimmune disorders and immune-related toxicities after immune checkpoint blockade for cancer. A confirmed role for the TNFSF/TNFRSF members in treating cancer has proven more elusive. In this review, we highlight the cancer-relevant TNFSF/TNFRSF family members, focusing on the death domain-containing and co-stimulation members and their signaling pathways, as well as their complicated role in the life and death of cancer cells.
Collapse
Affiliation(s)
- Obada Ababneh
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Daisuke Nishizaki
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- WIN Consortium, Paris, France.
- Department of Medicine, MCW Cancer Center, Milwaukee, WI, USA.
- Department of Oncology, University of Nebraska, Omaha, NE, USA.
| |
Collapse
|
27
|
Licht P, Mailänder V. Multi-Omic Data Integration Suggests Putative Microbial Drivers of Aetiopathogenesis in Mycosis Fungoides. Cancers (Basel) 2024; 16:3947. [PMID: 39682136 DOI: 10.3390/cancers16233947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/16/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Mycosis fungoides (MF) represents the most prevalent entity of cutaneous T cell lymphoma (CTCL). The MF aetiopathogenesis is incompletely understood, due to significant transcriptomic heterogeneity and conflicting views on whether oncologic transformation originates in early thymocytes or mature effector memory T cells. Recently, using clinical specimens, our group showed that the skin microbiome aggravates disease course, mainly driven by an outgrowing, pathogenic S. aureus strain carrying the virulence factor spa, which was shown by others to activate the T cell signalling pathway NF-κB. METHODS To explore the role of the skin microbiome in MF aetiopathogenesis, we here performed RNA sequencing, multi-omic data integration of the skin microbiome and skin transcriptome using Multi-Omic Factor Analysis (MOFA), virome profiling, and T cell receptor (TCR) sequencing in 10 MF patients from our previous study group. RESULTS We observed that inter-patient transcriptional heterogeneity may be largely attributed to differential activation of T cell signalling pathways. Notably, the MOFA model resolved the heterogenous activation pattern of T cell signalling after denoising the transcriptome from microbial influence. The MOFA model suggested that the outgrowing S. aureus strain evoked signalling by non-canonical NF-κB and IL-1B, which in turn may have fuelled the aggravated disease course. Further, the MOFA model indicated aberrant pathways of early thymopoiesis alongside enrichment of antiviral innate immunity. In line with this, viral prevalence, particularly of Epstein-Barr virus (EBV), trended higher in both lesional skin and the blood compared to nonlesional skin. Additionally, TCRs in both MF skin lesions and the blood were significantly more likely to recognize EBV peptides involved in latent infection. CONCLUSIONS First, our findings suggest that S. aureus with its virulence factor spa fuels MF progression through non-canonical NF-κB and IL-1B signalling. Second, our data provide insights into the potential role of viruses in MF aetiology. Last, we propose a model of microbiome-driven MF aetiopathogenesis: Thymocytes undergo initial oncologic transformation, potentially caused by viruses. After maturation and skin infiltration, an outgrowing, pathogenic S. aureus strain evokes activation and maturation into effector memory T cells, resulting in aggressive disease. Further studies are warranted to verify and extend our data, which are based on computational analyses.
Collapse
Affiliation(s)
- Philipp Licht
- Department of Dermatology, University Medical Centre Mainz, 55131 Mainz, Germany
| | - Volker Mailänder
- Department of Dermatology, University Medical Centre Mainz, 55131 Mainz, Germany
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| |
Collapse
|
28
|
Guo Y, Xie F, Liu X, Ke S, Chen J, Zhao Y, Li N, Wang Z, Yi G, Shen Y, Li D, Zhu C, Zhang Z, Zhao G, Lu H, Li B, Zhao W. Blockade of TNF-α/TNFR2 signalling suppresses colorectal cancer and enhances the efficacy of anti-PD1 immunotherapy by decreasing CCR8+T regulatory cells. J Mol Cell Biol 2024; 16:mjad067. [PMID: 37935468 PMCID: PMC11587560 DOI: 10.1093/jmcb/mjad067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 05/05/2023] [Accepted: 11/02/2023] [Indexed: 11/09/2023] Open
Abstract
The enrichment of regulatory T cells (Tregs) in the tumour microenvironment (TME) has been recognized as one of the major factors in the initiation and development of resistance to immune checkpoint inhibitors. C-C motif chemokine receptor 8 (CCR8), a marker of activated suppressive Tregs, has a significant impact on the functions of Tregs in the TME. However, the regulatory mechanism of CCR8 in Tregs remains unclear. Here, we revealed that a high level of TNF-α in the colorectal cancer (CRC) microenvironment upregulated CCR8 expression in Tregs via the TNFR2/NF-κB signalling pathway and the FOXP3 transcription factor. Furthermore, in both anti-programmed cell death protein 1 (anti-PD1)-responsive and anti-PD1-unresponsive tumour models, PD1 blockade induced CCR8+ Treg infiltration. In both models, Tnfr2 depletion or TNFR2 blockade suppressed tumour progression by reducing CCR8+ Treg infiltration and thus augmented the efficacy of anti-PD1 therapy. Finally, we identified that TNFR2+CCR8+ Tregs but not total Tregs were positively correlated with adverse prognosis in patients with CRC and gastric cancer. Our work reveals the regulatory mechanisms of CCR8 in Tregs and identifies TNFR2 as a promising target for immunotherapy.
Collapse
Affiliation(s)
- Yixian Guo
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Feng Xie
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xu Liu
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shouyu Ke
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jieqiong Chen
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yi Zhao
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Ning Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine Shanghai 200025, China
| | - Zeyu Wang
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Gang Yi
- Biotheus Inc., Zhuhai 519080, China
| | - Yanying Shen
- Department of Pathology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Dan Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chunchao Zhu
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zizhen Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Gang Zhao
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hong Lu
- GI Division, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bin Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wenyi Zhao
- Department of Gastrointestinal Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
29
|
Xiong W, Li Y, Hu L, He G, Huang J. Risks of malignancies related to disease-modifying antirheumatic drugs in rheumatoid arthritis: a pharmacovigilance analysis using the FAERS database. Front Pharmacol 2024; 15:1458500. [PMID: 39605908 PMCID: PMC11598350 DOI: 10.3389/fphar.2024.1458500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Objectives Over the years when disease-modifying antirheumatic drugs (DMARDs) have been used in rheumatoid arthritis patients, reports of malignancies have emerged. This study aims to investigate the association between malignancies and DMARDs by using data extracted from the Food and Drug Administration Adverse Event Reporting System (FAERS). Methods FAERS data (January 2019 to December 2023) were reviewed. For each drug-event pair, the disproportionality analysis was conducted to evaluate the risk of malignancy. Multivariate logistic regression was implemented to mitigate potential biases. Moreover, the time to onset of malignancy was also evaluated. Results We conducted a detailed search for rheumatoid arthritis indications and identified a total of 17,412 adverse event reports associated with malignancies, with selective DMARDs designated as the role code "primary suspect". At the preferred term level, there were 198 positive signals, among which the lower limit of the 95% confidence interval for the information component is 3.55 for squamous cell carcinoma of the skin, 2.39 for breast cancer, and 2.27 for lymphoproliferative disorder. In comparison to other DMARDs, targeted synthetic DMARDs were associated with a broader range of malignancies at both preferred term and Standardized MedDRA Queries levels. The number of adverse events reported in female patients is approximately 2-3 times higher than men, and the median age across the population was approximately 62 years. In terms of onset time, the conventional synthetic DMRADs exhibited a relatively longer median time, ranging from 3.58 to 7.08 years, while the targeted synthetic DMARDs demonstrated a shorter median time of 0.83-1.67 years. Conclusion Our study uncovers varying degrees of malignancy risks related to DMARDs, with a significantly higher risk observed in targeted synthetic DMARDs. Additionally, novel malignancy signals, not documented in product labels, have been detected. In the future, further research will be necessary to validate our findings.
Collapse
Affiliation(s)
- Wan Xiong
- Department of Pharmacy, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pharmacy, The First Hospital of Changsha, Changsha, China
| | - Yilin Li
- Department of Information and Digital Technology, PowerChina Zhongnan Engineering Corporation Limited, Changsha, China
| | - Lin Hu
- Department of Pharmacy, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pharmacy, The First Hospital of Changsha, Changsha, China
| | - Gefei He
- Department of Pharmacy, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pharmacy, The First Hospital of Changsha, Changsha, China
| | - Juanjuan Huang
- Department of Pharmacy, The Affiliated Changsha Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Department of Pharmacy, The First Hospital of Changsha, Changsha, China
| |
Collapse
|
30
|
Gao X, Li Y, Wang C, Zhao G. TIPE2 inhibits melanoma progression through MEK/ERK signaling. Sci Rep 2024; 14:27736. [PMID: 39532931 PMCID: PMC11557980 DOI: 10.1038/s41598-024-76794-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
Recent studies have uncovered that TIPE2 is involved in the development of cancer. However, less research has been conducted on the role of TIPE2 in melanoma. Our study aims to elucidate the mechanism of action of TIPE2 in the development of melanoma. We examined TIPE2 expression in paracarcinoma tissue and melanoma tissues and found that TIPE2 expression was downregulated in melanoma tissue compared with paracarcinoma tissue. Overexpression of TIPE2 significantly inhibited the proliferation of melanoma cells in vitro and even inhibited tumor formation in vivo. The CCK8 assay results indicated that TIPE2 overexpression suppressed the proliferation of melanoma cells. The colony-forming ability and wound healing ability of TIPE2-overexpressing melanoma cells were significantly reduced compared with those of control cells. Moreover, immunohistochemistry experiments using a nude mouse tumor model showed consistent results. TIPE2 inhibited the phosphorylation of MEK and ERK. In summary, TIPE2 suppresses the proliferation and migration of melanoma cells by affecting proliferation-related factors and possibly by regulating the MEK/ERK pathway. TIPE2 could be used to inhibit melanoma growth and is a potential drug target for future drug development.
Collapse
Affiliation(s)
- Xin Gao
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yan Li
- Department of Dermatology, ZiBo Central Hospital, No. 54 Gongqingtuan West Road, Zibo, 255000, Shandong, China
| | - Congcong Wang
- Department of Dermatology, ZiBo Central Hospital, No. 54 Gongqingtuan West Road, Zibo, 255000, Shandong, China
| | - Guowei Zhao
- Department of Dermatology, ZiBo Central Hospital, No. 54 Gongqingtuan West Road, Zibo, 255000, Shandong, China.
| |
Collapse
|
31
|
Sharma Y, Bala K. Multifarious Aspect of Cytokines as an Immuno-Therapeutic for Various Diseases. J Interferon Cytokine Res 2024; 44:477-485. [PMID: 39394036 DOI: 10.1089/jir.2024.0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024] Open
Abstract
Cytokines are known to be a group of growing small proteins that are majorly responsible for the transmission of signals and communication between hematopoietic cells, the cells of the human immune system, and other types of cells. Cytokines play a dominant role in different types of disorders and in perpetuating the inflammation-related disorders. The production of cytokines is a natural process inside the body of a human being against any foreign invasion or due to some pathogenic state to maintain the homeostasis. Cytokines respond in two ways; in some cases, the production and development of cytokines as a therapeutic discovery or intervention will enhance the treatment process and support the reaction given by the body against any pathogenic activity, and in some cases, overproduction of these cytokines responds in the opposite way and behaves as antagonists toward a typical therapeutic drug and its treatment. Overall, 41 articles were reviewed, and it was found that cytokines have proved to be a therapeutic approach among various diseases and can be utilized as a good candidate or a better choice for cancer therapeutics in future development.
Collapse
Affiliation(s)
- Yash Sharma
- Department of Biotechnology, IILM University, Greater Noida, India
| | - Kumud Bala
- Department of Biotechnology, IILM University, Greater Noida, India
- Therapeutics and Molecular Diagnostic Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, India
| |
Collapse
|
32
|
Fathima F, Subramaniyan Y, Rai A, Rekha PD. Enterococcus faecalis co-cultured with oral cancer cells exhibits higher virulence and promotes cancer cell survival, proliferation, and migration: an in vitro study. J Med Microbiol 2024; 73. [PMID: 39585322 DOI: 10.1099/jmm.0.001931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024] Open
Abstract
Introduction. Enterococcus faecalis is a common pathogen associated with many oral diseases and is often isolated from oral cancer patients. However, limited information is available on its key virulence gene expression in oral cancer cell microenvironment and cancer cell behaviour in co-culture studies.Hypothesis. E. faecalis overexpresses virulence genes when co-cultured with oral cancer cells and possibly alters the tumour microenvironment, promoting oral cancer proliferation and survival.Aim. To investigate altered virulence gene expression in E. faecalis and oral cancer cell behaviour using in vitro co-culture experiments.Methodology. Cal27 cells were co-cultured with E. faecalis and assessed for their cell proliferation, apoptosis, migration and clonogenicity using standard cell culture assays. The levels of reactive oxygen species (ROS) and inflammatory cytokines, along with proliferative, angiogenic and apoptotic biomarker expressions, were also assessed. E. faecalis adherence to cancer cells was demonstrated by the gentamicin protection assay. Real time-PCR was used to analyse the expression of virulence genes.Results. Co-culture of Cal27 cells with E. faecalis showed significantly higher cell proliferation, migration and clonogenicity compared to the control (P<0.01). A significant increase in the levels of ROS and inflammatory cytokines and overexpression of Ki67, vascular endothelial growth factor, extracellular signal-regulated kinase 1/2, phosphoinositide 3 kinase and Akt was observed in the co-culture group. E. faecalis also downregulated p53 and Bax genes while upregulated Bcl-2. The virulence genes GelE, Asa and Ace were overexpressed in E. faecalis co-cultured with Cal27 cells.Conclusion. The results from this study indicate the possible risks of E. faecalis infection in oral cancer. An effective antibiotic strategy against E. faecalis to prevent complications associated with oral diseases, including cancer, is needed.
Collapse
Affiliation(s)
- Fida Fathima
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Yuvarajan Subramaniyan
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Akshatha Rai
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Punchappady Devasya Rekha
- Division of Microbiology and Biotechnology, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| |
Collapse
|
33
|
Skapinker E, Aucoin EB, Kombargi HL, Yaish AM, Li Y, Baghaie L, Szewczuk MR. Contemporaneous Inflammatory, Angiogenic, Fibrogenic, and Angiostatic Cytokine Profiles of the Time-to-Tumor Development by Cancer Cells to Orchestrate Tumor Neovascularization, Progression, and Metastasis. Cells 2024; 13:1739. [PMID: 39451257 PMCID: PMC11506673 DOI: 10.3390/cells13201739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Cytokines can promote various cancer processes, such as angiogenesis, epithelial to mesenchymal transition (EMT), invasion, and tumor progression, and maintain cancer stem-cell-like (CSCs) cells. The mechanism(s) that continuously promote(s) tumors to progress in the TME still need(s) to be investigated. The data in the present study analyzed the inflammatory, angiogenic, fibrogenic, and angiostatic cytokine profiles in the host serum during tumor development in a mouse model of human pancreatic cancer. Pancreatic MiaPaCa-2-eGFP cancer cells were subcutaneously implanted in RAG2xCγ double mutant mice. Blood samples were collected before cancer cell implantation and every week until the end point of the study. The extracted serum from the blood of each mouse at different time points during tumor development was analyzed using a Bio-Plex microarray analysis and a Bio-Plex 200 system for proinflammatory (IL-1β, IL-10, IFN-γ, and TNF-α) and angiogenic and fibrogenic (IL-15, IL-18, basic FGF, LIF, M-CSF, MIG, MIP-2, PDGF-BB, and VEGF) cytokines. Here, we find that during cancer cell colonization for tumor development, host angiogenic, fibrogenic, and proinflammatory cytokine profiling in the tumor-bearing mice has been shown to significantly reduce host angiostatic and proinflammatory cytokines that restrain tumor development and increase those for tumor growth. The proinflammatory cytokines IL-15, IL-18, and IL-1β profiles reveal a significant host serum increase after day 35 when the tumor began to progress in growth. In contrast, the angiostatic cytokine profiles of TNFα, MIG, M-CSF, IL-10, and IFNγ in the host serum revealed a dramatic and significant decrease after day 5 post-implantation of cancer cells. OP treatment of tumor-bearing mice on day 35 maintained high levels of angiostatic and fibrogenic cytokines. The data suggest an entirely new regulation by cancer cells for tumor development. The findings identify for the first time how pancreatic cancer cells use host cytokine profiling to orchestrate the initiation of tumor development.
Collapse
Affiliation(s)
- Elizabeth Skapinker
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | - Emilyn B. Aucoin
- Faculty of Science, Biology (Biomedical Science), York University, Toronto, ON M3J 1P3, Canada;
| | - Haley L. Kombargi
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (H.L.K.); (A.M.Y.)
| | - Abdulrahman M. Yaish
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada; (H.L.K.); (A.M.Y.)
| | - Yunfan Li
- Faculty of Arts and Science, Queen’s University, Kingston, ON K7L 3N9, Canada; (E.S.); (Y.L.)
| | - Leili Baghaie
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| | - Myron R. Szewczuk
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada;
| |
Collapse
|
34
|
Toumi HR, Sallabi SM, Lubbad L, Al-Salam S, Hammad FT. The Effect of Nerolidol on Renal Dysfunction following Bilateral Ureteral Obstruction. Biomedicines 2024; 12:2285. [PMID: 39457599 PMCID: PMC11505435 DOI: 10.3390/biomedicines12102285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: Obstructive uropathy is a common cause of renal impairment. Recently, there has been a burgeoning interest in exploring natural products as potential alternative remedies for many conditions due to their low toxicity, affordability and wide availability. Methods: We investigated the effect of nerolidol in a rat model of bilateral ureteral obstruction (BUO) injury. Nerolidol, dissolved in a vehicle, was administered orally as a single daily dose of 200 mg/kg to Wistar rats. Sham group (n = 12) underwent sham surgery, whereas the BUO (n = 12) and BUO/NR groups (n = 12) underwent reversible 24-h BUO and received the vehicle or nerolidol, respectively. The treatment started 9 days prior to the BUO/sham surgery and continued for 3 days after reversal. Renal functions were assessed before starting the treatment, just prior to the intervention and 3 days after BUO reversal. Results: Neither nerolidol nor the vehicle affected the basal renal functions. Nerolidol resulted in a significant attenuation in the BUO-induced alterations in renal functional parameters such as serum creatinine and urea, creatinine clearance and urinary albumin-creatinine ratio. Nerolidol also attenuated the changes in several markers associated with renal injury, inflammation, apoptosis and oxidative stress and mitigated the histological alterations. Conclusions: The findings of this study demonstrated the potent reno-protective effects of nerolidol in mitigating the adverse renal effects of bilateral ureteral obstruction. This is attributed to its anti-inflammatory, anti-fibrotic, anti-apoptotic and anti-oxidant properties. These effects were reflected in the partial recovery of renal functions and histological features. These findings may have potential therapeutic implications.
Collapse
Affiliation(s)
- Harun R. Toumi
- Department of Surgery, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (H.R.T.); (S.M.S.); (L.L.)
| | - Sundus M. Sallabi
- Department of Surgery, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (H.R.T.); (S.M.S.); (L.L.)
| | - Loay Lubbad
- Department of Surgery, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (H.R.T.); (S.M.S.); (L.L.)
| | - Suhail Al-Salam
- Department of Pathology, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates;
| | - Fayez T. Hammad
- Department of Surgery, College of Medicine & Health Sciences, United Arab Emirates University, Al Ain P.O. Box 17666, United Arab Emirates; (H.R.T.); (S.M.S.); (L.L.)
| |
Collapse
|
35
|
Mansour HMM, Shehata MG, Darwish AMG, Hafez EE, Samy MA, Abdelmotilib NM, Abdo EM. Antioxidant and anti-cancer potentials of Ag green-synthesized and encapsulated olive leaves particles on HCT-116 cells. Int J Biol Macromol 2024; 278:134776. [PMID: 39153672 DOI: 10.1016/j.ijbiomac.2024.134776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 08/13/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Water extracts (OLE), whey protein encapsulated extracts (OLE/WPNs), and silver nanoparticles (OLE/Ag-NPs) were prepared from olive leaves of Manzenllie and Picual varieties. These preparations were characterized, and their antioxidant and biological activities on Vero and HCT-116 colorectal cells were assessed. The mechanism of action of the preparations was studied through tumor necrosis factor-α (TNF-α) and cytochrome C oxidase (Cox1) gene expression. OLE/Ag-NPs showed smaller particle sizes (14.23-15.53 nm) than OLE/WPNs (229.83-310.67 nm) and demonstrated lower aggregation due to their high Ƹ-potential of -24.86 to -27.90 mV. None of the preparations affected the viability of Vero cells (IC50 = 192.19-421.01 μg/mL), but they showed cytotoxic effects on HCT-116 cells (IC50 = 50.76-196.54 μg/mL), particularly OLE/WPNs. Moreover, the preparations from the Picual variety (OLE, OLE/WPNs, and OLE/Ag-NPs) showed regulatory effects against colon cancer on treated HCT-116 cells by upregulating Cox1 expression and downregulating TNF-α expression. Consequently, OLE/WPNs and OLE/Ag-NPs could be promising for industrial applications with potential health benefits.
Collapse
Affiliation(s)
- Hanem M M Mansour
- Food Technology Department, Arid Lands Cultivation Research Institute (ALCRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El Arab, P.O. Box 21934, Alexandria, Egypt
| | - Mohamed G Shehata
- Food Technology Department, Arid Lands Cultivation Research Institute (ALCRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El Arab, P.O. Box 21934, Alexandria, Egypt; Food Research Section, R&D Division, Abu Dhabi Agriculture and Food Safety Authority (ADAFSA), Abu Dhabi, P.O. Box 52150, United Arab Emirates
| | - Amira M G Darwish
- Food Technology Department, Arid Lands Cultivation Research Institute (ALCRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El Arab, P.O. Box 21934, Alexandria, Egypt; Food Industry Technology Program, Faculty of Industrial and Energy Technology, Borg Al Arab Technological University (BATU), Alexandria, Egypt.
| | - Elsayed Elsayed Hafez
- Plant Protection and Bio-Molecular Diagnosis Department, Arid Lands Cultivation Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El Arab, P.O. Box 21934, Alexandria, Egypt
| | - Marwa A Samy
- Plant Protection and Bio-Molecular Diagnosis Department, Arid Lands Cultivation Research Institute, City of Scientific Research and Technological Applications (SRTA-City), New Borg El Arab, P.O. Box 21934, Alexandria, Egypt
| | - Neveen M Abdelmotilib
- Food Technology Department, Arid Lands Cultivation Research Institute (ALCRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El Arab, P.O. Box 21934, Alexandria, Egypt
| | - Eman M Abdo
- Food Science Department, Faculty of Agriculture (Saba Basha), Alexandria University, P.O. Box 21531, Alexandria, Egypt.
| |
Collapse
|
36
|
Santi MD, Zhang M, Asam K, Yu G, Dong PM, Sheehan DH, Aouizerat BE, Thomas CM, Viet CT, Ye Y. Perineural Invasion Is Associated With Function-evoked Pain and Altered Extracellular Matrix in Patients With Head and Neck Squamous Cell Carcinoma. THE JOURNAL OF PAIN 2024; 25:104615. [PMID: 38936749 DOI: 10.1016/j.jpain.2024.104615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 06/01/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is painful, and perineural invasion (PNI) has been associated with the worst pain. Pain due to HNSCC is diverse and may vary based on clinicopathological factors. This study aims to characterize different pain patterns linked with PNI, its influence on daily functioning, and gain insights into molecular changes and pathways associated with PNI-related pain in HNSCC patients. We conducted a cross-sectional study across 3 medical centers (n = 114), assessing pain phenotypes and their impact on daily functioning using 2 self-reported pain questionnaires, given to patients prior to their cancer surgery. Furthermore, we conducted RNA-seq analysis utilizing the The Cancer Genome Atlas dataset of HNSCC tumor from patients (n = 192) to identify genes relevant to both PNI and pain. Upon adjusting for demographic and clinicopathological variables using linear regression models, we found that PNI independently predicted function-evoked pain according to the University of Calfornia San Francisco Oral Cancer Pain Questionnaire, as well as the worst pain intensity reported in the Brief Pain Inventory. Distinct pain patterns were observed to be associated with daily activities in varying manners. Our molecular analyses revealed significant disruptions in pathways associated with the extracellular matrix structure and organization. The top differentially expressed genes linked to the extracellular matrix are implicated in cancer development, pain, and neurodegenerative diseases. Our data underscore the importance of properly categorizing pain phenotypes in future studies aiming to uncover mechanistic underpinnings of pain. Additionally, we have compiled a list of genes of interest that could serve as targets for both cancer and cancer pain management. PERSPECTIVE: PNI independently predicts function-evoked pain. Different pain phenotypes affect daily activities differently. We identified a list of candidate genes involved in the extracellular matrix structure and function that can be targeted for both cancer and cancer pain control.
Collapse
Affiliation(s)
- Maria D Santi
- Translational Research Center, College of Dentistry, New York University, New York, New York; Department of Molecular Pathobiology, Pain Research Center, College of Dentistry, New York University, New York, New York
| | - Morgan Zhang
- Translational Research Center, College of Dentistry, New York University, New York, New York; Department of Molecular Pathobiology, Pain Research Center, College of Dentistry, New York University, New York, New York
| | - Kesava Asam
- Translational Research Center, College of Dentistry, New York University, New York, New York
| | - Gary Yu
- Rory Meyers College of Nursing, New York University, New York, New York
| | - Phuong M Dong
- Department of Oral and Maxillofacial Surgery, Loma Linda University School of Dentistry, Loma Linda, California
| | - Delaney H Sheehan
- Department of Otolaryngology - Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bradley E Aouizerat
- Translational Research Center, College of Dentistry, New York University, New York, New York
| | - Carissa M Thomas
- Department of Otolaryngology - Head & Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Chi T Viet
- Department of Oral and Maxillofacial Surgery, Loma Linda University School of Dentistry, Loma Linda, California
| | - Yi Ye
- Translational Research Center, College of Dentistry, New York University, New York, New York; Department of Molecular Pathobiology, Pain Research Center, College of Dentistry, New York University, New York, New York.
| |
Collapse
|
37
|
Meidenbauer J, Wachter M, Schulz SR, Mostafa N, Zülch L, Frey B, Fietkau R, Gaipl US, Jost T. Inhibition of ATM or ATR in combination with hypo-fractionated radiotherapy leads to a different immunophenotype on transcript and protein level in HNSCC. Front Oncol 2024; 14:1460150. [PMID: 39411143 PMCID: PMC11473424 DOI: 10.3389/fonc.2024.1460150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024] Open
Abstract
Background The treatment of head and neck tumors remains a challenge due to their reduced radiosensitivity. Small molecule kinase inhibitors (smKI) that inhibit the DNA damage response, may increase the radiosensitivity of tumor cells. However, little is known about how the immunophenotype of the tumor cells is modulated thereby. Therefore, we investigated whether the combination of ATM or ATR inhibitors with hypo-fractionated radiotherapy (RT) has a different impact on the expression of immune checkpoint markers (extrinsic), the release of cytokines or the transcriptome (intrinsic) of head and neck squamous cell carcinoma (HNSCC) cells. Methods The toxic and immunogenic effects of the smKI AZD0156 (ATMi) and VE-822 (ATRi) in combination with a hypo-fractionated scheme of 2x5Gy RT on HPV-negative (HSC4, Cal-33) and HPV-positive (UM-SCC-47, UD-SCC-2) HNSCC cell lines were analyzed as follows: cell death (necrosis, apoptosis; detected by AnxV/PI), expression of immunostimulatory (ICOS-L, OX40-L, TNFSFR9, CD70) and immunosuppressive (PD-L1, PD-L2, HVEM) checkpoint marker using flow cytometry; the release of cytokines using multiplex ELISA and the gene expression of Cal-33 on mRNA level 48 h post-RT. Results Cell death was mainly induced by the combination of RT with both inhibitors, but stronger with ATRi. Further, the immune phenotype of cancer cells, not dying from combination therapy itself, is altered predominantly by RT+ATRi in an immune-stimulatory manner by the up-regulation of ICOS-L. However, the analysis of secreted cytokines after treatment of HNSCC cell lines revealed an ambivalent influence of both inhibitors, as we observed the intensified secretion of IL-6 and IL-8 after RT+ATRi. These findings were confirmed by RNAseq analysis and further the stronger immune-suppressive character of RT+ATMi was enlightened. We detected the down-regulation of a central protein of cytoplasmatic sensing pathways of nucleic acids, RIG-1, and found one immune-suppressive target, EDIL3, strongly up-regulated by RT+ATMi. Conclusion Independent of a restrictive toxicity, the combination of RT + either ATMi or ATRi leads to comprehensive and immune-modulating alterations in HNSCC. This includes pro-inflammatory signaling induced by RT + ATRi but also anti-inflammatory signals. These findings were confirmed by RNAseq analysis, which further highlighted the immune-suppressive nature of RT + ATMi.
Collapse
Affiliation(s)
- Julia Meidenbauer
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Wachter
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian R. Schulz
- Division of Molecular Immunology, Internal Medicine III, University Hospital Erlangen, Nikolaus-Fiebiger Center, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Nada Mostafa
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Lilli Zülch
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Benjamin Frey
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Uniklinikum Erlangen, Erlangen, Germany
- FAU Profile Center Immunomedicine Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Rainer Fietkau
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Uniklinikum Erlangen, Erlangen, Germany
- FAU Profile Center Immunomedicine Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Udo S. Gaipl
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Uniklinikum Erlangen, Erlangen, Germany
- FAU Profile Center Immunomedicine Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tina Jost
- Translational Radiobiology, Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Radiation Oncology, Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-Europäische Metropolregion Nürnberg (EMN), Uniklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
38
|
Huang D, Tu Z, Karnoub AE, Wei W, Rezaeian AH. Busulfan Chemotherapy Downregulates TAF7/TNF-α Signaling in Male Germ Cell Dysfunction. Biomedicines 2024; 12:2220. [PMID: 39457533 PMCID: PMC11504710 DOI: 10.3390/biomedicines12102220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/07/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Busulfan is an FDA-approved alkylating drug used in the chemotherapy of advanced acute myeloid leukemia. The precise mechanisms by which Busulfan kills spermatogonia stem cells (SSCs) are not yet completely understood. Methods: Using a murine model, we evaluated Busulfan-induced apoptosis and DNA damage signaling between testis and ovary tissues. We executed RT-qPCR, analyzed single-nuclei RNA sequencing data and performed in situ hybridization for the localization of the gene expression in the tissues. Results: The results indicate that, in contrast to female germ cells, haploid male germ cells undergo significant apoptosis following Busulfan chemotherapy. Moreover, a gene enrichment analysis revealed that reactive oxygen species may activate the inflammatory response in part through the TNF-α/NF-κB signaling pathway. Interestingly, in the testis, the mRNA levels of TNF-α and TAF7 (TATA box-binding protein-associated factor 7) are downregulated, and testosterone levels suppressed. Mechanistically, the promoter of TNF-α has a conserved motif for binding TAF7, which is necessary for its transcriptional activation and may require further in-depth study. We next analyzed the tumorigenic function of TAF7 and revealed that it is highly overexpressed in several types of human cancers, particularly testicular germ cell tumors, and associated with poor patient survival. Therefore, we executed in situ hybridization and single-nuclei RNA sequencing, finding that less TAF7 mRNA is present in SSCs after chemotherapy. Conclusions: Thus, our data indicate a possible function of TAF7 in the regulation of SSCs and spermatogenesis following downregulation by Busulfan. These findings may account for the therapeutic effects of Busulfan and underlie its potential impact on cancer chemotherapy prognosis.
Collapse
Affiliation(s)
| | | | | | | | - Abdol-Hossein Rezaeian
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
39
|
Tsoupras A, Adamantidi T, Finos MA, Philippopoulos A, Detopoulou P, Tsopoki I, Kynatidou M, Demopoulos CA. Re-Assessing the Role of Platelet Activating Factor and Its Inflammatory Signaling and Inhibitors in Cancer and Anti-Cancer Strategies. FRONT BIOSCI-LANDMRK 2024; 29:345. [PMID: 39473406 DOI: 10.31083/j.fbl2910345] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/14/2024] [Accepted: 07/03/2024] [Indexed: 01/03/2025]
Abstract
Since 2000s, we have outlined the multifaceted role of inflammation in several aspects of cancer, via specific inflammatory mediators, including the platelet activating factor (PAF) and PAF-receptor (PAFR) related signaling, which affect important inflammatory junctions and cellular interactions that are associated with tumor-related inflammatory manifestations. It is now well established that disease-related unresolved chronic inflammatory responses can promote carcinogenesis. At the same time, tumors themselves are able to promote their progression and metastasis, by triggering an inflammation-related vicious cycle, in which PAF and its signaling play crucial role(s), which usually conclude in tumor growth and angiogenesis. In parallel, new evidence suggests that PAF and its signaling also interact with several inflammation-related cancer treatments by inducing an antitumor immune response or, conversely, promoting tumor recurrence. Within this review article, the current knowledge and future perspectives of the implication of PAF and its signaling in all these important aspects of cancer are thoroughly re-assessed. The potential beneficial role of PAF-inhibitors and natural or synthetic modulators of PAF-metabolism against tumors, tumor progression and metastasis are evaluated. Emphasis is given to natural and synthetic molecules with dual anti-PAF and anti-cancer activities (Bio-DAPAC-tives), with proven evidence of their antitumor potency through clinical trials, as well as on metal-based anti-inflammatory mediators that constitute a new class of potent inhibitors. The way these compounds may promote anti-tumor effects and modulate the inflammatory cellular actions and immune responses is also discussed. Limitations and future perspectives on targeting of PAF, its metabolism and receptor, including PAF-related inflammatory signaling, as part(s) of anti-tumor strategies that involve inflammation and immune response(s) for an improved outcome, are also evaluated.
Collapse
Affiliation(s)
- Alexandros Tsoupras
- Hephaestus Laboratory, School of Chemistry, Faculty of Science, Democritus University of Thrace, Kavala University Campus, 65404 Kavala, Greece
| | - Theodora Adamantidi
- Hephaestus Laboratory, School of Chemistry, Faculty of Science, Democritus University of Thrace, Kavala University Campus, 65404 Kavala, Greece
| | - Marios Argyrios Finos
- Hephaestus Laboratory, School of Chemistry, Faculty of Science, Democritus University of Thrace, Kavala University Campus, 65404 Kavala, Greece
| | - Athanassios Philippopoulos
- Laboratory of Inorganic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Paraskevi Detopoulou
- Department of Nutritional Sciences and Dietetics, University of the Peloponnese, 24100 Kalamata, Greece
| | - Ifigeneia Tsopoki
- Department of Dietetics and Nutrition, Harokopio University, 17676 Kallithea, Greece
| | - Maria Kynatidou
- Department of Dietetics and Nutrition, Harokopio University, 17676 Kallithea, Greece
| | - Constantinos A Demopoulos
- Laboratory of Biochemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece
| |
Collapse
|
40
|
Mahooti M, Abdolalipour E, Sanami S, Zare D. Inflammatory Modulation Effects of Probiotics: A Safe and Promising Modulator for Cancer Prevention. Curr Microbiol 2024; 81:372. [PMID: 39312034 DOI: 10.1007/s00284-024-03901-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 09/15/2024] [Indexed: 10/22/2024]
Abstract
Chronic inflammation is the gate of many human illnesses and happens when the immune system is unable to suppress external attacks in the correct form. Nonetheless, the gut microbiome plays a pivotal role in keeping homeostasis in the human body and preventing inflammation. Imbalanced microbiota and many diseases can result in inflammation, which when not taken seriously, can be turned into chronic ones and ultimately lead to serious diseases such as cancer. One approach to maintaining hemostasis in the human body is consumption of probiotics as a supplement. Probiotics impact the immune functions of dendritic cells (DCs), T cells, and B cells in the gut-associated lymphoid tissue by inducing the secretion of an array of cytokines. They activate the innate immune response through their microbial-associated molecular pattern, and this activation is followed by multiple cytokine secretion and adaptive elicitation that mitigates pro-inflammatory expression levels and tumor incidence. Thus, according to several studies showing the benefit of probiotics application, alone or in combination with other agents, to induce potent immune responses in individuals against some inflammatory disorders and distinct types of cancers, this review is devoted to surveying the role of probiotics and the modulation of inflammation in some cancer models.
Collapse
Affiliation(s)
- Mehran Mahooti
- Department of Biotechnology, Iranian Research Organization for Science and Technology, P. O. Box 3353-5111, Tehran, Iran
| | - Elahe Abdolalipour
- Department of Virology, Pasteur Institute of Iran, P.O.Box: 1316943551, Tehran, Iran
| | - Samira Sanami
- Ubnormal Uterine Bleeding Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Davood Zare
- Department of Biotechnology, Iranian Research Organization for Science and Technology, P. O. Box 3353-5111, Tehran, Iran.
| |
Collapse
|
41
|
Zhang S, Yang Y, Zhang L, Liu Y, Guo Z, Wu J, Zhou W, Hong Z, Zhang W. Identification and Validation of a Prognostic Signature Based on Fibroblast Immune-related Genes to Predict the Prognosis and Therapeutic Response of renal clear cell carcinoma by Integrated Analysis of Single-Cell and Bulk RNA Sequencing Data. J Cancer 2024; 15:5942-5955. [PMID: 39440053 PMCID: PMC11493018 DOI: 10.7150/jca.100194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 08/24/2024] [Indexed: 10/25/2024] Open
Abstract
Background: The importance of fibroblasts in cancer progression is becoming more acknowledged, particularly the significance of their immune-related genes. However, the precise roles these genes play in fibroblasts throughout tumor development remains unclear. Exploring how these genes function in advancing kidney renal clear cell carcinoma (KIRC) could provide answers to these uncertainties. Material and method: The Cancer Genome Atlas (TCGA) database served as the source of data for KIRC patients. We distinguished fibroblast immune-related genes (FIGs), which are used to construct risk score. Further analysis conducted including enrichment analysis, assessment of tumor mutation burden (TMB), evaluation of tumor microenvironment (TME), analysis of immune cell infiltration, and drug sensitivity prediction. Result: The risk score using 6 FIGs effectively predicts the outcomes for KIRC patients. Nomogram which is based on the risk score and clinical data, demonstrated superior predictive performance compared to the version without the risk score. Enrichment analysis identified that coagulation pathway predominates in high-risk group, the protein secretion pathway is prevalent in low-risk patients' cohort. The adverse prognosis in high-risk patient cohort could be linked to an elevated TMB. TME analysis showed that high-risk group's tumor tissues contain more immune and stromal cells. Furthermore, the amount of regulatory T cells increases with the risk score. Low-risk group response better to immunotherapy. Finally, RT-qPCR confirmed the differential expression of FIGs in KIRC patients. Conclusion: This risk score and nomogram are valuable tools assessing KIRC patients' prognosis. Poorer prognosis in high-risk categories may have relationship with activation of coagulation pathway and a higher TMB.
Collapse
Affiliation(s)
- Shuwen Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
- Queen Mary School, Jiangxi medical college, Nanchang University, Nanchang, China, 330006
| | - Yuqian Yang
- Queen Mary School, Jiangxi medical college, Nanchang University, Nanchang, China, 330006
- Department of Urology Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Liyi Zhang
- Queen Mary School, Jiangxi medical college, Nanchang University, Nanchang, China, 330006
- Department of Urology Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Yijiang Liu
- Department of Urology Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
- Jiangxi medical college, Nanchang University, Nanchang, China, 330006
| | - Zihun Guo
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Jiajun Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Weijun Zhou
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Zhengdong Hong
- Department of Urology Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| | - Wenxiong Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China, 330006
| |
Collapse
|
42
|
Dzikowiec M, Galant S, Lik P, Góralska K, Nejc D, Piekarski J, Majos A, Brzeziańska-Lasota E, Pastuszak-Lewandoska D. Analysis of Spermine Oxidase gene and proinflammatory cytokines expression in gastric cancer patients with and without Helicobacter pylori infection - A pilot study in Polish population. Adv Med Sci 2024; 69:443-450. [PMID: 39305951 DOI: 10.1016/j.advms.2024.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/09/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024]
Abstract
PURPOSE Many types of cancer have infectious origins. Gastric cancer patients can demonstrate high seroprevalence of Helicobacter pylori (H. pylori). The aim of the present study was to assess the expression of SMOX gene in the group of Polish patients with gastric cancer. SMOX is believed to promote H. pylori-induced carcinogenesis via inflammation, DNA damage and activation of β-catenin signaling. We also assessed the mRNA expression of selected pro-inflammatory cytokines, i.e. IL-2, IFN-γ, TNF-α, and antimicrobial peptide, cathelicidin. MATERIALS/METHODS The study material consisted of gastric tissue samples collected during total gastrectomy from three different places in stomach: from primary tumor, 3 cm away from the primary lesion, and from the wall opposite to the primary tumor. After RNA isolation, qPCR reactions were performed for the relevant genes. RESULTS The obtained results confirmed an increased level of SMOX expression in gastric cancer patients with the history of H. pylori infection. And, as far as we know, this is the first study on SMOX gene expression conducted on tissue taken from a patient, not on a cell line. The levels of pro-inflammatory cytokines, i.e. IL-2, IFN-γ, TNF-α, were also increased, thus indicating their contribution to the specific inflammatory microenvironment of the tumor. Interestingly, the levels of CAMP, encoding antimicrobial peptide, were reduced in all tissue types. CONCLUSIONS The findings confirm that SMOX plays a role in gastric carcinogenesis. However, further research is needed on the role of inflammatory and other factors involved in this process to identify targets for cancer immunotherapy.
Collapse
Affiliation(s)
- Magdalena Dzikowiec
- Department of Biology and Parasitology, Biology and Medical Microbiology, Medical University of Lodz, Lodz, Poland.
| | - Sandra Galant
- Department of Biology and Parasitology, Biology and Medical Microbiology, Medical University of Lodz, Lodz, Poland
| | - Przemysław Lik
- Department of Surgical Oncology, Medical University of Lodz, Lodz, Poland
| | - Katarzyna Góralska
- Department of Biology and Parasitology, Biology and Medical Microbiology, Medical University of Lodz, Lodz, Poland
| | - Dariusz Nejc
- Department of Surgical Oncology, Medical University of Lodz, Lodz, Poland
| | - Janusz Piekarski
- Department of Surgical Oncology, Medical University of Lodz, Lodz, Poland
| | - Alicja Majos
- Department of General and Transplant Surgery, Medical University of Lodz, Lodz, Poland
| | - Ewa Brzeziańska-Lasota
- Department of Biomedicine and Genetics, Biology and Medical Microbiology, Medical University of Lodz, Lodz, Poland
| | - Dorota Pastuszak-Lewandoska
- Department of Microbiology and Laboratory Medical Immunology, Biology and Medical Microbiology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
43
|
Coënon L, Geindreau M, Ghiringhelli F, Villalba M, Bruchard M. Natural Killer cells at the frontline in the fight against cancer. Cell Death Dis 2024; 15:614. [PMID: 39179536 PMCID: PMC11343846 DOI: 10.1038/s41419-024-06976-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 07/31/2024] [Accepted: 08/05/2024] [Indexed: 08/26/2024]
Abstract
Natural Killer (NK) cells are innate immune cells that play a pivotal role as first line defenders in the anti-tumor response. To prevent tumor development, NK cells are searching for abnormal cells within the body and appear to be key players in immunosurveillance. Upon recognition of abnormal cells, NK cells will become activated to destroy them. In order to fulfill their anti-tumoral function, they rely on the secretion of lytic granules, expression of death receptors and production of cytokines. Additionally, NK cells interact with other cells in the tumor microenvironment. In this review, we will first focus on NK cells' activation and cytotoxicity mechanisms as well as NK cells behavior during serial killing. Lastly, we will review NK cells' crosstalk with the other immune cells present in the tumor microenvironment.
Collapse
Affiliation(s)
- Loïs Coënon
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
| | - Mannon Geindreau
- Equipe TIRECs, Labellisée Ligue Contre le Cancer, Centre de Recherche INSERM CTM-UMR1231, Dijon, France
- University of Bourgogne Franche-Comté, Dijon, France
| | - François Ghiringhelli
- Equipe TIRECs, Labellisée Ligue Contre le Cancer, Centre de Recherche INSERM CTM-UMR1231, Dijon, France
- University of Bourgogne Franche-Comté, Dijon, France
- Platform of Transfer in Biological Oncology, Georges-François Leclerc Cancer Center, Dijon, France
| | - Martin Villalba
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, Montpellier, France
- Institut du Cancer Avignon-Provence Sainte Catherine, Avignon, France
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Mélanie Bruchard
- Equipe TIRECs, Labellisée Ligue Contre le Cancer, Centre de Recherche INSERM CTM-UMR1231, Dijon, France.
- University of Bourgogne Franche-Comté, Dijon, France.
- Platform of Transfer in Biological Oncology, Georges-François Leclerc Cancer Center, Dijon, France.
| |
Collapse
|
44
|
Ahuja S, Lazar IM. Proteomic insights into breast cancer response to brain cell-secreted factors. Sci Rep 2024; 14:19351. [PMID: 39169222 PMCID: PMC11339284 DOI: 10.1038/s41598-024-70386-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/16/2024] [Indexed: 08/23/2024] Open
Abstract
The most devastating feature of cancer cells is their ability to metastasize to distant sites in the body. HER2 + and TN breast cancers frequently metastasize to the brain and stay potentially dormant for years until favorable conditions support their proliferation. The sheltered and delicate nature of the brain prevents, however, early disease detection and effective delivery of therapeutic drugs. Moreover, the challenges associated with the acquisition of brain biopsies add compounding difficulties to exploring the mechanistic aspects of tumor development. To provide insights into the determinants of cancer cell behavior at the brain metastatic site, this study was aimed at exploring the early response of HER2 + breast cancer cells (SKBR3) to factors present in the brain perivascular niche. The neural microenvironment was simulated by using the secretome of a set of brain cells that come first in contact with the cancer cells upon crossing the blood brain barrier, i.e., endothelial cells, astrocytes, and microglia. Cytokine microarrays were used to investigate the secretome mediators of intercellular communication, and proteomic technologies for assessing the changes in the behavior of cancer cells upon exposure to the brain cell-secreted factors. The cytokines detected in the brain secretomes were supportive of inflammatory conditions, while the SKBR3 cells secreted numerous cancer-promoting growth factors that were either absent or present in lower abundance in the brain cell cultures, indicating that upon exposure the SKBR3 cells may have been deprived of favorable conditions for optimal growth. Altogether, the results suggest that the exposure of SKBR3 cells to the brain cell-secreted factors altered their growth potential and drove them toward a state of quiescence, with broader overall outcomes that affected cellular metabolism, adhesion and immune response processes. The findings of this study underscore the key role played by the neural niche in shaping the behavior of metastasized cancer cells, provide insights into the cellular cross-talk that may lead cancer cells into dormancy, and highlight novel opportunities for the development of metastatic breast cancer therapeutic strategies.
Collapse
Affiliation(s)
- Shreya Ahuja
- Department of Biological Sciences, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA
| | - Iulia M Lazar
- Department of Biological Sciences, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA.
- Fralin Life Sciences Institute, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA.
- Carilion School of Medicine, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA.
- Division of Systems Biology/AIS, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA, 24061, USA.
| |
Collapse
|
45
|
Wang KC, Sun YS, Tsai HC, Liao HT, Lai CC, Chen WS, Lu LY, Chen MH. Increased risk of malignancy in HLA-B27-positive patients with ankylosing spondylitis requiring biologics for sustained inflammation: A long-term, single-center retrospective study. Mod Rheumatol 2024; 34:1027-1035. [PMID: 38302085 DOI: 10.1093/mr/roae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 01/10/2024] [Indexed: 02/03/2024]
Abstract
OBJECTIVES To assess the link between the administration of biologic disease-modifying antirheumatic drugs (bDMARDs) and the risk of malignancy in human leukocyte antigen B27 (HLA-B27)-positive patients with ankylosing spondylitis (AS) experiencing sustained inflammation. METHODS Between 2006 and 2021, 1445 HLA-B27-positive patients with AS were retrospectively evaluated. Among them, 112 patients required bDMARD therapy. The study compared conventional therapy with bDMARDs and investigated the risk factors for developing malignancies. RESULTS During 8253 patient-years of follow-up, 38 (2.6%) patients developed various malignancies, including lung, liver, breast, and colon cancer. The risk of malignancy was significantly higher in the bDMARD-treated group compared to PS-matched groups receiving conventional synthetic DMARDs (csDMARD) and non-steroidal anti-inflammatory drugs. The cumulative risk of malignancies increased significantly after 6 years of follow-up. All patients who developed malignancy after bDMARD therapy received tumor necrosis factor-α inhibitors. Requiring bDMARD therapy, requiring bDMARDs in combination with csDMARD therapy, and being diagnosed with AS after 30 years of age were independent risk factors for developing malignancy. CONCLUSIONS HLA-B27-positive AS patients with sustained inflammation requiring biologic therapy, particularly if diagnosed after age 30, may have an increased risk of malignancy. Regular cancer screenings are advisable for these patients while undergoing biologic treatment.
Collapse
Affiliation(s)
- Kai-Chun Wang
- Division of Allergy-Immunology-Rheumatology, Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- The Doctoral Program of Clinical and Experimental Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Yi-Syuan Sun
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hung-Cheng Tsai
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsien-Tzung Liao
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chien-Chih Lai
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Sheng Chen
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ling-Ying Lu
- Division of Allergy-Immunology-Rheumatology, Department of Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
- School of Medicine, College of Medicine, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Ming-Han Chen
- Division of Allergy, Immunology and Rheumatology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Faculty of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| |
Collapse
|
46
|
Zhao Q, Yu H, Shi M, Wang X, Fan Z, Wang Z. Tumor microenvironment characteristics of lipid metabolism reprogramming related to ferroptosis and EndMT influencing prognosis in gastric cancer. Int Immunopharmacol 2024; 137:112433. [PMID: 38870879 DOI: 10.1016/j.intimp.2024.112433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/05/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Gastric cancer (GC) is a refractory malignant tumor with high tumor heterogeneity, a low rate of early diagnosis, and poor patient prognosis. Lipid metabolism reprogramming plays a critical role in tumorigenesis and progression, but its prognostic role and regulatory mechanism in GC are rarely studied. Thus, the identification of signatures related to lipid metabolism is necessary and may present a new avenue for improving the overall prognosis of GC. METHODS Lipid metabolism-associated genes (LMAGs) with differential expression in tumor and tumor-adjacent tissue were acquired to identify lipid metabolism-associated subtypes. The differentially expressed genes (DEGs) between the two clusters were then utilized for prognostic analysis and signature construction. Additionally, pathway enrichment analysis and immune cell infiltration analysis were employed to identify the characteristics of the prognostic model. Further analyses were conducted at the single-cell level to better understand the model's prognostic mechanism. Finally, the prediction of immunotherapy response was used to suggest potential treatments. RESULTS Two lipid metabolism-associated subtypes were identified and 9 prognosis-related genes from the DEGs between the two clusters were collected for the construction of the prognostic model named lipid metabolism-associated signature (LMAS). Then we found the low LMAS patients with favorable prognoses were more sensitive to ferroptosis in the Cancer Genome Atlas of Stomach Adenocarcinoma (TCGA-STAD). Meanwhile, the tumor cells exhibiting high levels of lipid peroxidation and accumulation of reactive oxygen species (ROS) in single-cell levels were primarily enriched in the low LMAS group, which was more likely to induce ferroptosis. In addition, endothelial cells and cancer-associated fibroblasts (CAFs) facilitated tumor angiogenesis, proliferation, invasion, and metastasis through endothelial-mesenchymal transition (EndMT), affecting the prognosis of the patients with high LMAS scores. Moreover, CD1C- CD141- dendritic cells (DCs) also secreted pro-tumorigenic cytokines to regulate the function of endothelial cells and CAFs. Finally, the patients with low LMAS scores might have better efficacy in immunotherapy. CONCLUSIONS A LMAS was constructed to guide GC prognosis and therapy. Meanwhile, a novel anti-tumor effect was found in lipid metabolism reprogramming of GC which improved patients' prognosis by regulating the sensitivity of tumor cells to ferroptosis. Moreover, EndMT may have a negative impact on GC prognosis.
Collapse
Affiliation(s)
- Qian Zhao
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou 014030, China; School of Basic Medicine, Baotou Medical College, Baotou 014040, China
| | - Hui Yu
- Translational Medicine Center, Baotou Medical College, Baotou 014040, China
| | - Mengqi Shi
- School of Basic Medicine, Baotou Medical College, Baotou 014040, China
| | - Xujie Wang
- School of Basic Medicine, Baotou Medical College, Baotou 014040, China
| | - Zixu Fan
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou 014030, China
| | - Zhanli Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou 014030, China.
| |
Collapse
|
47
|
Rogovskii V. Tumor-produced immune regulatory factors as a therapeutic target in cancer treatment. Front Immunol 2024; 15:1416458. [PMID: 39206193 PMCID: PMC11349530 DOI: 10.3389/fimmu.2024.1416458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Affiliation(s)
- Vladimir Rogovskii
- Department of Molecular Pharmacology and Radiobiology, Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
48
|
Duan R, Milton P, Sittplangkoon C, Liu X, Sui Z, Boyce BF, Yao Z. Chimeric antigen receptor dendritic cells targeted delivery of a single tumoricidal factor for cancer immunotherapy. Cancer Immunol Immunother 2024; 73:203. [PMID: 39105847 PMCID: PMC11303651 DOI: 10.1007/s00262-024-03788-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T cells have been used to treat blood cancers by producing a wide variety of cytokines. However, they are not effective in treating solid cancers and can cause severe side-effects, including cytokine release syndrome. TNFα is a tumoricidal cytokine, but it markedly increases the protein levels of cIAP1 and cIAP2, the members of inhibitor of apoptosis protein (IAP) family of E3 ubiquitin ligase that limits caspase-induced apoptosis. Degradation of IAP proteins by an IAP antagonist does not effectively kill cancer cells but enables TNFα to strongly induce cancer cell apoptosis. It would be a promising approach to treat cancers by targeted delivery of TNFα through an inactive adoptive cell in combination with an IAP antagonist. METHODS Human dendritic cells (DCs) were engineered to express a single tumoricidal factor, TNFα, and a membrane-anchored Mucin1 antibody scFv, named Mucin 1 directed DCs expressing TNFα (M-DCsTNF). The efficacy of M-DCsTNF in recognizing and treating breast cancer was tested in vitro and in vivo. RESULTS Mucin1 was highly expressed on the surface of a wide range of human breast cancer cell lines. M-DCsTNF directly associated with MDA-MB-231 cells in the bone of NSG mice. M-DCsTNF plus an IAP antagonist, SM-164, but neither alone, markedly induce MDA-MB-231 breast cancer cell apoptosis, which was blocked by TNF antibody. Importantly, M-DCsTNF combined with SM-164, but not SM-164 alone, inhibited the growth of patient-derived breast cancer in NSG mice. CONCLUSION An adoptive cell targeting delivery of TNFα combined with an IAP antagonist is a novel effective approach to treat breast cancer and could be expanded to treat other solid cancers. Unlike CAR-T cell, this novel adoptive cell is not activated to produce a wide variety of cytokines, except for additional overexpressed TNF, and thus could avoid the severe side effects such as cytokine release syndrome.
Collapse
Affiliation(s)
- Rong Duan
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Philip Milton
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
- School of Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Chutamath Sittplangkoon
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Xin Liu
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
- Department of Orthopedics, Tianjin Hospital, Tianjin, 30021, People's Republic of China
| | - Zhining Sui
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Brendan F Boyce
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA
| | - Zhenqiang Yao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA.
| |
Collapse
|
49
|
Saboorifar H, Zafarani Y, Gholampour G, Roghani A, Qiu F, Dequaniter D, Yu Q. Serum inflammatory markers as prognostic marker for nasopharyngeal carcinoma with liver metastasis: a multi-center retrospective study. Eur Arch Otorhinolaryngol 2024; 281:4315-4324. [PMID: 38642085 DOI: 10.1007/s00405-024-08649-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 03/28/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND This retrospective study investigated the prognostic value of serum inflammatory markers in nasopharyngeal carcinoma (NPC) patients, focusing on their association with overall survival (OS) and liver metastasis-free survival (LMFS). METHODS The study included 314 NPC patients treated between 2010 and 2020. Clinical characteristics, treatment methods, and serum inflammatory markers were assessed. Patients were categorized into two groups of with and without liver metastasis. Univariate and multivariate Cox regression and Kaplan-Meier survival analyses were performed to investigate the prognostic value of serum inflammatory markers in NPC patients with and without liver metastasis. RESULTS In the whole cohort, univariate Cox regression analysis singled out tumor necrosis factor-α (TNF-α) (HR = 1.57, 95% CI 1.44-4.90, p = 0.004) and neutrophil-to-lymphocyte ratio (NLR) (HR = 2.13, 95% CI 1.33-3.99, p = 0.009), which were significantly associated with poorer OS. In patients with liver metastasis, TNF-α and NLR could not independently predict OS. However, high TNF-α levels were independently associated with worse OS in patients without liver metastasis (HR (95% CI) = 2.75 (1.67-8.68), p < 0.001). High NLR levels could independently predict poor OS in both groups with (HR (95% CI) = 1.94 (1.77-6.38), p = 0.010) and without liver metastasis (HR (95% CI) = 1.58 (1.19-7.54), p = 0.009). Ultimately, TNF-α and NLR could not significantly predict LMFS. CONCLUSION This study highlights the prognostic significance of TNF-α and NLR in NPC patients, especially in those with liver metastasis. These inflammatory markers could serve as valuable indicators for assessing the prognosis of NPC patients. Further research is warranted to validate their clinical utility and explore potential therapeutic implications.
Collapse
Affiliation(s)
- Hossein Saboorifar
- Department of Head and Neck Surgery, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Yasamin Zafarani
- Department of Head and Neck Surgery, School of Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Golsa Gholampour
- Department of Head and Neck Surgery, Tehran University of Medical Sciences, Tehran, Iran
| | - Arman Roghani
- Department of Head and Neck Surgery, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Feng Qiu
- Department of Radiation Oncology, Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Dideir Dequaniter
- Medicine Faculty, Université Libre de Bruxelles (ULB), Route de Lennik 808, Brussels, Belgium
| | - Qiao Yu
- Otorhinolaryngology and Maxillofacial Department, First Affiliated Hospital of Nanchang University, Nanchang, China.
| |
Collapse
|
50
|
Abdala-Díaz RT, Casas-Arrojo V, Castro-Varela P, Riquelme C, Carrillo P, Medina MÁ, Cárdenas C, Becerra J, Pérez Manríquez C. Immunomodulatory, Antioxidant, and Potential Anticancer Activity of the Polysaccharides of the Fungus Fomitiporia chilensis. Molecules 2024; 29:3628. [PMID: 39125036 PMCID: PMC11314378 DOI: 10.3390/molecules29153628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/15/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Fomitiporia species have aroused the interest of numerous investigations that reveal their biological activity and medicinal potential. The present investigation shows the antioxidant, anticancer, and immunomodulatory activity of acidic polysaccharides obtained from the fungus Fomitiporia chilensis. The acidic polysaccharides were obtained for acidic precipitation with 2% O-N-cetylpyridinium bromide. Chemical analysis was performed using FT-IR and GC-MS methods. The antioxidant capacity of acidic polysaccharides from F. chilensis was evaluated by scavenging free radicals with an ABTS assay. Macrophage proliferation and cytokine production assays were used to determine the immunomodulatory capacity of the polysaccharides. Anti-tumor and cytotoxicity activity was evaluated with an MTT assay in the U-937, HTC-116, and HGF-1 cell lines. The effect of polysaccharides on the cell cycle of the HCT-116 cell line was determined for flow cytometry. Fourier Transform-infrared characterization revealed characteristic absorption peaks for polysaccharides, whereas the GC-MS analysis detected three peaks corresponding to D-galactose, galacturonic acid, and D-glucose. The secreted TNF-α concentration was increased when the cell was treated with 2 mg mL-1 polysaccharides, whereas the IL-6 concentration was increased with all of the evaluated polysaccharide concentrations. A cell cycle analysis of HTC-116 treated with polysaccharides evidenced that the acidic polysaccharides from F. chilensis induce an increase in the G0/G1 cell cycle phase, increasing the apoptotic cell percentage. Results from a proteomic analysis suggest that some of the molecular mechanisms involved in their antioxidant and cellular detoxifying effects and justify their traditional use in heart diseases. Proteomic data are available through ProteomeXchange under identifier PXD048361. The study on acidic polysaccharides from F. chilensis has unveiled their diverse biological activities, including antioxidant, anticancer, and immunomodulatory effects. These findings underscore the promising therapeutic applications of acidic polysaccharides from F. chilensis, warranting further pharmaceutical and medicinal research exploration.
Collapse
Affiliation(s)
- Roberto T. Abdala-Díaz
- Department of Ecology and Geology, Institute of Blue Biotechnology and Development (IBYDA), Malaga University, E-29071 Malaga, Spain; (R.T.A.-D.); (V.C.-A.)
| | - Virginia Casas-Arrojo
- Department of Ecology and Geology, Institute of Blue Biotechnology and Development (IBYDA), Malaga University, E-29071 Malaga, Spain; (R.T.A.-D.); (V.C.-A.)
| | - Pablo Castro-Varela
- FICOLAB Microalgal Research Group, Department of Botany, Faculty of Natural and Oceanographic Sciences, University of Concepción, Concepción PC 304000, Chile;
| | - Cristian Riquelme
- Mycology Laboratory, Institute of Biochemistry and Microbiology, Universidad Austral de Chile, Isla Teja, PO 567, Valdivia PC 5049000, Chile;
| | - Paloma Carrillo
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Andalucía Tech, E-29071 Málaga, Spain; (P.C.); (M.Á.M.); (C.C.)
- Malaga Biomedical Research Institute and Nanomedicine Platform (IBIMA PlataformaBIONAND), C/Severo Ochoa, 35, E-29590 Málaga, Spain
| | - Miguel Ángel Medina
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Andalucía Tech, E-29071 Málaga, Spain; (P.C.); (M.Á.M.); (C.C.)
- Malaga Biomedical Research Institute and Nanomedicine Platform (IBIMA PlataformaBIONAND), C/Severo Ochoa, 35, E-29590 Málaga, Spain
- Network Biomedical Research Center for Rare Diseases (CIBERER), U741, E-28029 Málaga, Spain
| | - Casimiro Cárdenas
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, University of Málaga, Andalucía Tech, E-29071 Málaga, Spain; (P.C.); (M.Á.M.); (C.C.)
- Research Support Central Services (SCAI) of the University of Málaga, E-29071 Málaga, Spain
| | - José Becerra
- Laboratory of Chemistry of Natural Products, Department of Botany, Faculty of Natural and Oceanographic Sciences, University of Concepción, Concepción PC 304000, Chile;
- Technological Development Unit, University of Concepción, Concepción PC 304000, Chile
| | - Claudia Pérez Manríquez
- Laboratory of Chemistry of Natural Products, Department of Botany, Faculty of Natural and Oceanographic Sciences, University of Concepción, Concepción PC 304000, Chile;
- Technological Development Unit, University of Concepción, Concepción PC 304000, Chile
| |
Collapse
|