1
|
Rastogi I, Mannone JA, Gibadullin R, Moseman JE, Sidney J, Sette A, McNeel DG, Gellman SH. β-amino acid substitution in the SIINFEKL antigen alters immunological recognition. Cancer Biol Ther 2025; 26:2486141. [PMID: 40200635 PMCID: PMC11988276 DOI: 10.1080/15384047.2025.2486141] [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: 02/18/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Peptide vaccines offer a direct way to initiate an immunogenic response to a defined antigen epitope. However, peptide vaccines are unstable in vivo, subject to rapid enzymatic proteolysis. Replacement of an α-amino acid residue with a homologous β-amino acid residue (native side chain, but backbone extended by a single CH2 unit) impairs proteolysis at nearby amide bonds. Therefore, antigen analogues containing α-to-β replacements have been examined for functional mimicry of native all-α antigens. Another group previously took this approach in the ovalbumin (OVA) antigen model by evaluating single α-to-β analogues of the murine major histocompatibility complex (MHC) I-restricted peptide SIINFEKL. METHODS We re-examined this set of α/β SIINFEKL antigens. We tested the susceptibility to proteolysis in mouse serum and their ability to activate OVA-antigen-specific CD8 T cells in vitro. Additionally, we tested the α/β antigens in vivo for their ability to induce an antigen-specific immunogenic response in naïve mice and in OVA-expressing tumor-bearing mice. RESULTS The α/β antigens were comparable to the native antigen in their susceptibility to proteolysis in serum. Each α/β antigen was capable of activating antigen-specific CD8 T cells in vitro. However, antigen-specific CD8 T cells induced against α/β antigens in vivo were not cross-reactive to the native antigen. Moreover, immunization with α/β analogues did not elicit anti-tumor effects in tumor-bearing mice. CONCLUSIONS We conclude that even though α/β analogues of the SIINFEKL antigen can elicit a T cell-based response, this class of backbone-modified peptides is not promising from the perspective of antitumor vaccine development.
Collapse
Affiliation(s)
- Ichwaku Rastogi
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - John A. Mannone
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Ruslan Gibadullin
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Jena E. Moseman
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, University of California, San Diego, CA, USA
| | - Douglas G. McNeel
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Samuel H. Gellman
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
2
|
Piao YH, Luo J, Ren T, Wang A, Li YY, Pan LJ, Li XZ, Li FT, Bao YW, Zheng F, Yue H. Integrating Intestinal Biotransformation and Gut Microbiota to Uncover the Influence of Tongfu Xiexia Decoction in Rats With Constipation. RAPID COMMUNICATIONS IN MASS SPECTROMETRY : RCM 2025; 39:e10065. [PMID: 40329014 DOI: 10.1002/rcm.10065] [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: 02/16/2025] [Revised: 04/24/2025] [Accepted: 04/27/2025] [Indexed: 05/08/2025]
Abstract
RATIONALE Tongfu Xiexia Decoction (TFXXD), a traditional Chinese medicine formula, comprises six herbs: semen raphani, Rehmannia glutinosa, rhubarb, Magnolia officinalis, trifoliate orange, and mirabilite. The aim of this study was to investigate the effects of TFXXD on its biotransformation and microbial abundance in the gut of constipated rats. METHODS Ultrahigh-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UHPLC-Q-TOF-MS/MS) was used to analyze the biotransformation of TFXXD in the colonic contents, and 16S rRNA sequencing was used to assess the structure and diversity of the gut microbiota across various rat groups. RESULTS We identified and analyzed 25 biotransformations of TFXXD in the colonic contents, which undergo various reactions such as deglycosylation, ring opening, and hydration in intestinal bacteria, with 14 originating from trifoliate orange, six from rhubarb, three from Rehmannia glutinosa, one from Semen raphani, and one from M. officinalis. 16S rRNA analysis revealed that TFXXD significantly enhanced the relative abundance of beneficial bacteria, such as Lactobacillus and Bacteroides while significantly reducing Oscillospira abundance. Moreover, TFXXD considerably affected the carbohydrate and amino acid metabolism. Correlation analyses revealed a significant negative correlation between Bacteroides and ACH (Acetylcholine), NO (nitric oxide), D-Lac (D-Lactate), IL-6 (Interleukin-6), TNF-α (tumor necrosis factor-α), and IL-1β (Interleukin-1β) and a significant positive correlation between Bacteroides, hesperetin, and rhein. CONCLUSIONS In conclusion, our findings indicate that TFXXD can modulate the structure and diversity of the gut microbiota and enhance the metabolic balance in constipated rats.
Collapse
Affiliation(s)
- Yu-Han Piao
- Changchun University of Chinese Medicine, Changchun, China
| | - Jing Luo
- Changchun University of Chinese Medicine, Changchun, China
| | - Tao Ren
- Changchun University of Chinese Medicine, Changchun, China
| | - Ao Wang
- Changchun University of Chinese Medicine, Changchun, China
| | - Yu-Yun Li
- Jilin City Hospital of Chemical Industry, Jilin, China
| | - Li-Jia Pan
- Changchun University of Chinese Medicine, Changchun, China
| | - Xin-Ze Li
- Changchun University of Chinese Medicine, Changchun, China
| | - Fang-Tong Li
- Changchun University of Chinese Medicine, Changchun, China
| | - Yu-Wen Bao
- Changchun University of Chinese Medicine, Changchun, China
| | - Fei Zheng
- Changchun University of Chinese Medicine, Changchun, China
| | - Hao Yue
- Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
3
|
Cui X, Liu W, Jiang H, Zhao Q, Hu Y, Tang X, Liu X, Dai H, Rui H, Liu B. IL-12 family cytokines and autoimmune diseases: A potential therapeutic target? J Transl Autoimmun 2025; 10:100263. [PMID: 39759268 PMCID: PMC11697604 DOI: 10.1016/j.jtauto.2024.100263] [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: 10/20/2024] [Revised: 11/28/2024] [Accepted: 12/01/2024] [Indexed: 01/07/2025] Open
Abstract
In recent years, the discovery of IL-12 family cytokines, which includes IL-12, IL-23, IL-27, IL-35, and IL-39, whose biological functions directly or indirectly affect various autoimmune diseases. In autoimmune diseases, IL-12 family cytokines are aberrantly expressed to varying degrees. These cytokines utilize shared subunits to influence T-cell activation and differentiation, thereby regulating the balance of T-cell subsets, which profoundly impacts the onset and progression of autoimmune diseases. In such conditions, IL-12 family members are aberrantly expressed to varying degrees. By exploring their immunomodulatory functions, researchers have identified varying therapeutic potentials for each member. This review examines the physiological functions of the major IL-12 family members and their interactions, discusses their roles in several autoimmune diseases, and summarizes the progress of clinical studies involving monoclonal antibodies targeting IL-12 and IL-23 subunits currently available for treatment.
Collapse
Affiliation(s)
- Xiaoyu Cui
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
| | - Wu Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Hanxue Jiang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
| | - Qihan Zhao
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
| | - Yuehong Hu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
| | - Xinyue Tang
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
| | - Xianli Liu
- Shunyi Branch, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100310, China
| | - Haoran Dai
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Shunyi Branch, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100310, China
| | - Hongliang Rui
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Beijing Institute of Chinese Medicine, Beijing, 100010, China
| | - Baoli Liu
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, 100010, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
4
|
Zununi Vahed S, Hejazian SM, Bakari WN, Landon R, Gueguen V, Meddahi-Pellé A, Anagnostou F, Barzegari A, Pavon-Djavid G. Milking mesenchymal stem cells: Updated protocols for cell lysate, secretome, and exosome extraction, and comparative analysis of their therapeutic potential. Methods 2025; 238:40-60. [PMID: 40058715 DOI: 10.1016/j.ymeth.2025.03.004] [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: 11/05/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/21/2025] Open
Abstract
The potential of the cell lysate, secretome, and extracellular vesicles (EVs) of mesenchymal stem cells (MSCs) to modulate the immune response and promote tissue regeneration has positioned them as a promising option for cell-free therapy. Currently, many clinical trials in stem cells-derived EVs and secretome are in progress various diseases and sometimes the results are failing. The major challenge on this roadmap is the lack of a standard extraction method for exosome, secretome, and lysate. The most optimal method for obtaining the secretome of MSCs for clinical utilization involves a comprehensive approach that includes non-destructive collection methods, time optimization, multiple collection rounds, optimization of culture conditions, and quality control measures. Further research and clinical studies are warranted to validate and refine these methods for safe and effective utilization of the MSC exosome, secretome, and lysate in various clinical applications. To address these challenges, it is imperative to establish a standardized and unified methodology to ensure reliable evaluation of these extractions in clinical trials. This review seeks to outline the pros and cons of methods for the preparation of MSCs-derived exosome, and secretome/lysate, and comparative analysis of their therapeutic potential.
Collapse
Affiliation(s)
| | | | - William Ndjidda Bakari
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France; Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Rebecca Landon
- Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Virginie Gueguen
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France
| | - Anne Meddahi-Pellé
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France
| | - Fani Anagnostou
- Université Paris Cité, CNRS UMR7052, INSERM U1271, ENVA, B3OA, F-75010 Paris, France
| | - Abolfazl Barzegari
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran; Research Center for Pharmaceutical Nanotechnology (RCPN), Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Graciela Pavon-Djavid
- Université Sorbonne Paris Nord, INSERM U1148, Laboratory for Vascular Translational Science, Nanotechnologies for Vascular Medicine and Imaging Team, 99 Av. Jean-Baptiste Clément 93430 Villetaneuse, France.
| |
Collapse
|
5
|
Wijfjes Z, Ramos Tomillero I, Le Gall CM, van Dinther EAW, Turlings F, Classens R, Manna S, van Dalen D, Peters RJRW, Schouren K, Fennemann FL, Hagemans IM, van Dalen FJ, van der Schoot JMS, Figdor CG, Esser-Kahn A, Scheeren FA, Verdoes M. Co-delivery of antigen and adjuvant by site-specific conjugation to dendritic cell-targeted Fab fragments potentiates T cell responses. RSC Chem Biol 2025:d5cb00014a. [PMID: 40343174 PMCID: PMC12057635 DOI: 10.1039/d5cb00014a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
The aim of therapeutic cancer vaccines is to induce tumor-specific cellular immune responses. This requires tumor antigens to be efficiently processed and presented by antigen-presenting cells, in particular dendritic cells (DCs). In addition, DCs require maturation to upregulate the surface expression and secretion of T cell costimulatory molecules, which is achieved by co-administration of adjuvants in vaccines. Peptide-based antigen vaccination is an attractive strategy due to the established biocompatibility of peptides as well as the dosing control. To enhance the efficacy of peptide-based vaccines, antigens can be targeted to DCs. Antigen-adjuvant conjugates are known to enhance T cell activation by ensuring DC maturation upon antigen delivery. In this study, we aim to combine these two approaches in a single molecule, and present a DC-targeted antibody fragment-antigen-adjuvant (AAA)-conjugate. We generate the AAA-conjugate through a combination of site-specific sortase-mediated chemoenzymatic ligation and click chemistry. Ex vivo T cell activation assays show enhanced efficacy of the AAA-conjugate compared to non-adjuvanted control conjugates. The in vivo performance of the AAA-conjugate was suboptimal, which we hypothesize to be a consequence of the hydrophobic character of the conjugate. In vivo efficacy was rescued by co-administration of antibody fragment-antigen conjugates and antibody fragment-adjuvant conjugates, in which the antigen and adjuvant were separatedly delivered using two different DC-targeting molecules. In conclusion, this study provides a proof-of-concept for effective in vivo antigen-specific T cell activation by targeted delivery of both antigen and adjuvant to DCs in a single or separate molecule using site-specific protein engineering.
Collapse
Affiliation(s)
- Zacharias Wijfjes
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
- Institute for Chemical Immunology Nijmegen The Netherlands
| | - Iván Ramos Tomillero
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Camille M Le Gall
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Eric A W van Dinther
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Frederique Turlings
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
- IMAGINE! Consortium Nijmegen The Netherlands
| | - René Classens
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Saikat Manna
- Pritzker School of Molecular Engineering, University of Chicago Chicago USA
| | - Duco van Dalen
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Ruud J R W Peters
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Kayleigh Schouren
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Felix L Fennemann
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Iris M Hagemans
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
| | - Floris J van Dalen
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
- Institute for Chemical Immunology Nijmegen The Netherlands
| | | | - Carl G Figdor
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
- Institute for Chemical Immunology Nijmegen The Netherlands
| | - Aaron Esser-Kahn
- Pritzker School of Molecular Engineering, University of Chicago Chicago USA
| | - Ferenc A Scheeren
- Department of Dermatology, Leiden University Medical Center Leiden The Netherlands
| | - Martijn Verdoes
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen The Netherlands
- Institute for Chemical Immunology Nijmegen The Netherlands
- IMAGINE! Consortium Nijmegen The Netherlands
| |
Collapse
|
6
|
Dan L, Kang-Zheng L. Optimizing viral transduction in immune cell therapy manufacturing: key process design considerations. J Transl Med 2025; 23:501. [PMID: 40316943 PMCID: PMC12046913 DOI: 10.1186/s12967-025-06524-0] [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: 12/31/2024] [Accepted: 04/21/2025] [Indexed: 05/04/2025] Open
Abstract
Immune cell therapies have revolutionized the treatment of cancers, autoimmune disorders, and infectious diseases. A critical step in their manufacturing is viral transduction, which enables the delivery of therapeutic genes into immune cells. However, the complexity of this process presents significant challenges for optimization and scalability. This review provides a comprehensive analysis of viral transduction process in immune cell therapy manufacturing, highlighting key design considerations to support the development of safe, effective, and scalable production methods. Additionally, it examines current technological challenges in immune cell transduction and explores future innovations poised to advance the field.
Collapse
Affiliation(s)
- Liu Dan
- Bioprocessing Technology Institute BTI, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Singapore, 138668, Singapore.
| | - Lee Kang-Zheng
- Bioprocessing Technology Institute BTI, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Singapore, 138668, Singapore
| |
Collapse
|
7
|
El-Bahrawy NR, Hafez AAAEL, Elmekawy A, Salem M, Sarhan N, Morsy R. Development and characterization of nano-hydroxyapatite/gelatin/PVA/alginate-based multifunctional active scaffolds for bone regeneration: An in vitro and in vivo study. Int J Biol Macromol 2025; 307:141160. [PMID: 39965692 DOI: 10.1016/j.ijbiomac.2025.141160] [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: 07/17/2024] [Revised: 02/04/2025] [Accepted: 02/15/2025] [Indexed: 02/20/2025]
Abstract
Multifunctional porous bone scaffolds that combine reparative and therapeutic features are promising for bone tissue engineering applications. Therefore, we developed freeze-dried scaffolds based on the in situ synthesis of hydroxyapatite nanoparticles (HAp NPs) within a gelatin-polyvinyl alcohol (PVA)-alginate matrix using a co-precipitation method. Ceftazidime and 5-fluorouracil (5-FU) were used as drug models and were separately loaded into the fabricated scaffolds. The hybrid scaffolds exhibited an ultimate compressive strength of 1.1 MPa and flexible behavior favored for fitting irregular bone defects. 5-FU-loaded scaffolds showed higher bioactive potential within 3 days compared to ceftazidime-loaded scaffolds. The scaffolds exhibited a long-term degradation rate, and thereby prolonged release of ceftazidim and 5-FU for up to 28 days. 5-FU-loaded scaffolds showed excellent nearly equal antibacterial activity to ceftazidime-loaded scaffolds against Staphylococcus epidermidis and Escherichia coli strains. Osteosarcoma cell death was achieved by increased concentrations of ceftazidime and 5-FU treatment above 300 μg/mL and 250 μg/mL, respectively. The developed scaffolds displayed higher bone formation ability with better osteogenesis in a femoral rat bone defect model compared to the control sample. This work represents a promising solution for bone defect repair and provides insight into the development of multifunctional scaffolds for local chemotherapy and bone tissue engineering applications.
Collapse
Affiliation(s)
- Nadia R El-Bahrawy
- Biophysics Lab, Physics Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | | | - Ahmed Elmekawy
- Biophysics Lab, Physics Department, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Mohamed Salem
- Immunology and Biotechnology Unit, Zoology Department, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Naglaa Sarhan
- Histology Department, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
| | - Reda Morsy
- Biophysics Lab, Physics Department, Faculty of Science, Tanta University, Tanta 31527, Egypt
| |
Collapse
|
8
|
Yang C, Ma H, Liang Z, Zhuang Y, Hu L, Zhang K, Huang L, Li M, Zhang S, Zhen Y. Cyclic RGD modified dextran-quercetin polymer micelles for targeted therapy of breast cancer. Int J Biol Macromol 2025; 308:142272. [PMID: 40118409 DOI: 10.1016/j.ijbiomac.2025.142272] [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: 08/02/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 03/23/2025]
Abstract
Quercetin is a natural flavonoid found in many plants which has various pharmacological activities including antitumor effect. However, the poor water solubility and bioavailability limit the potential benefits of quercetin for patients. Thus, modifying quercetin structure and developing actively targeted drug delivery systems are extremely important for tumor precision therapy. Herein, polymer-drug conjugates dextran-quercetin (D-Q) and cRGD-dextran (R-D) were synthesized by grafting quercetin and polypeptide cRGDfk (Arg-Gly-Asp-(D-Phe)-Lys) to dextran. Then cRGD-modified dextran-quercetin polymer micelles (R-D-Q) were constructed by self-assembling of D-Q and R-D. R-D-Q micelles possessed appropriate particle size (133.4 nm), nearly neutral potential (8.14 mV) and excellent drug-loading efficiency (13.1 %) and achieved higher cytotoxicity, apoptosis induction and penetration to human breast cancer MCF-7 cells than the micelles unmodified with cRGD, which were ascribed to cRGD-integrin mediated transcytosis. R-D-Q micelles effectively suppressed tumor growth in tumor-bearing mice by delivering more quercetin throughout the tumor tissue. And R-D-Q micelles could promote the apoptosis of tumor cells by activating p38 and JNK signal pathways and suppressing ERK signal pathway. In addition, R-D-Q micelles had no damage to normal tissues of mice at therapeutic dose. These results indicate promising prospects for R-D-Q micelles as an effective drug delivery system against tumor.
Collapse
Affiliation(s)
- Chunpeng Yang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Huiling Ma
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ze Liang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Ying Zhuang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Litao Hu
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Kexin Zhang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Leixiao Huang
- College of Pharmacy, Dalian Medical University, Dalian 116044, China
| | - Min Li
- Molecular Imaging Precision Medical Collaborative Innovation Center, Shanxi Medical University, Taiyuan 030001, China; Department of Nuclear Medicine, The First Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan 030001, China.
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Bioresources Utilization of Ministry of Education, Dalian Minzu University, Dalian 116600, China.
| | - Yuhong Zhen
- College of Pharmacy, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
9
|
Qin S, Na J, Yang Q, Tang J, Deng Y, Zhong L. Advances in dendritic cell-based therapeutic tumor vaccines. Mol Immunol 2025; 181:113-128. [PMID: 40120558 DOI: 10.1016/j.molimm.2025.03.005] [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: 12/18/2024] [Revised: 02/09/2025] [Accepted: 03/11/2025] [Indexed: 03/25/2025]
Abstract
Dendritic cell-based therapeutic tumor vaccines are an active immunotherapy that has been commonly tried in the clinic,traditional treatment modalities for malignant tumors, such as surgery, radiotherapy and chemotherapy, have the disadvantages of high recurrence rates and side effects. The dendritic cell vaccination destroys cells from tumors by means of the patient's own system of immunity, a very promising treatment. However, due to the suppression of the tumor immune microenvironment, the difficulty of screening for optimal specific antigens, and the high technical difficulty of vaccine production. Most tumor vaccines currently available in the clinic have failed to produce significant clinical therapeutic effects. In this review, the fundamentals of therapeutic dendritic cells vaccine therapy are briefly outlined, with a focus on the progress of therapeutic Dendritic cells vaccine research in the clinic and the initiatives undertaken to enhance dendritic cell vaccinations' anti-tumor effectiveness. It is believed that through the continuous exploration of novel therapeutic strategies, therapeutic dendritic cells vaccines can play a greater role in improving tumor treatment for tumor patients.
Collapse
Affiliation(s)
- Simin Qin
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Therangstics, Guangxi Key Laboratory of Bio-targeting Therangstics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China.
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Therangstics, Guangxi Key Laboratory of Bio-targeting Therangstics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China.
| | - Qun Yang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Therangstics, Guangxi Key Laboratory of Bio-targeting Therangstics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China.
| | - Jing Tang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Therangstics, Guangxi Key Laboratory of Bio-targeting Therangstics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China.
| | - Yamin Deng
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Therangstics, Guangxi Key Laboratory of Bio-targeting Therangstics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China.
| | - Liping Zhong
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Therangstics, Guangxi Key Laboratory of Bio-targeting Therangstics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
10
|
Liang MS, Huang Y, Huang SF, Zhao Q, Chen ZS, Yang S. Flavonoids in the Treatment of Non-small Cell Lung Cancer via Immunomodulation: Progress to Date. Mol Diagn Ther 2025; 29:307-327. [PMID: 40036006 DOI: 10.1007/s40291-025-00772-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2025] [Indexed: 03/06/2025]
Abstract
Lung cancer is one of the most common malignancies in the world, while non-small cell lung cancer (NSCLC) accounts for about 80% of all lung cancers. Most patients with NSCLC have advanced stage disease at diagnosis, and the 5-year survival rate can be discouragingly low. Flavonoids are widely found in fruits, vegetables, teas, and medicinal plants, with a variety of functional effects, including anti-inflammatory, antioxidant, and anticancer properties. This review aims to focus on the research progress of flavonoids in the treatment of NSCLC, including immunomodulatory effects on NSCLC, promotion of reactive oxygen species (ROS) production, interaction with microRNA (miRNA), and interactions with certain proteins. In addition, combining flavonoids and anticancer agents, radiotherapy, or nanoparticles can reverse NSCLC drug resistance, inducing apoptosis of cancer cells. It therefore appears that flavonoids alone or in combination with other treatment agents may be a promising therapeutic modality for treating NSCLC, with great potential in mass production and clinical applications.
Collapse
Affiliation(s)
- Man-Shan Liang
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yang Huang
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Sheng-Feng Huang
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Qi Zhao
- Cancer Center, Institute of Translational Medicine,Faculty of Health Sciences, University of Macau, Macau SPR, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SPR, China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY, USA.
| | - Shuo Yang
- Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, Department of Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
11
|
Canelo-Vilaseca M, Sabbah M, Di Blasi R, Cristinelli C, Sureda A, Caillat-Zucman S, Thieblemont C. Lymphodepletion chemotherapy in chimeric antigen receptor-engineered T (CAR-T) cell therapy in lymphoma. Bone Marrow Transplant 2025; 60:559-567. [PMID: 40148484 DOI: 10.1038/s41409-025-02539-9] [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: 08/10/2024] [Revised: 09/29/2024] [Accepted: 02/17/2025] [Indexed: 03/29/2025]
Abstract
The development of chimeric antigen receptor (CAR) T-cells, engineered from peripheral T-lymphocytes of a patient with lymphoma, in order to specifically target tumor cells, has been a revolution in adoptive cell therapy (ACT). As outlined in this review, ACT was initiated by hematopoietic cell transplantation (HSCT) and re-injection of interleukin-boosted tumor-infiltrating lymphocytes (TIL). The innovative venture of genetically modifying autologous peripheral T-cells to target them to cell-surface tumoral antigens through an antibody-derived structure (i.e. independent of major histocompatibility antigen presentation, physiologically necessary for T-cell activation), and intracytoplasmic T-cell costimulatory peptides, via a novel membrane CAR, has been an outstanding breakthrough. Here, focusing on B-cell hematological malignancies and mostly non-Hodgkin lymphoma, attention is brought to the importance of providing an optimal microenvironment for such therapeutic cells to proliferate and positively develop anti-tumoral cytotoxicity. This, perhaps paradoxically, implies a pre-infusion step of deep lymphopenia and deregulation of immunosuppressive mechanisms enhanced by tumoral cells. Fludarabine and cyclophosphamide appear to be the most efficient lymphodepletive drugs in this context, dosage being of importance, as will be illustrated by a thorough literature review.
Collapse
Affiliation(s)
- Marta Canelo-Vilaseca
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France
| | - Mohamad Sabbah
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France
- Université Paris Cité, Paris, France
| | - Roberta Di Blasi
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France
| | - Caterina Cristinelli
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d'Oncologia-L'Hospitalet, IDIBELL, Universitat de Barcelona, Barcelona, Spain
| | - Sophie Caillat-Zucman
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Laboratoire d'Immunologie, Paris, France
| | - Catherine Thieblemont
- Assistance Publique - Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Hémato-oncologie, Paris, France.
- Université Paris Cité, Paris, France.
- Inserm U1153, Hôpital Saint Louis, Paris, France.
| |
Collapse
|
12
|
Kapoor DU, Pareek A, Sharma S, Prajapati BG, Thanawuth K, Sriamornsak P. Alginate gels: Chemistry, gelation mechanisms, and therapeutic applications with a focus on GERD treatment. Int J Pharm 2025; 675:125570. [PMID: 40199431 DOI: 10.1016/j.ijpharm.2025.125570] [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: 01/07/2025] [Revised: 03/25/2025] [Accepted: 04/05/2025] [Indexed: 04/10/2025]
Abstract
Alginate, a natural polysaccharide derived primarily from marine algae, has become popular in biomedical research due to its versatile gelation properties and biocompatibility. This review explores the chemistry, gelation mechanisms, and therapeutic applications of alginate gels, with a particular focus on their role in gastroesophageal reflux disease (GERD) management. Alginate's structure, comprised of guluronic and mannuronic acid blocks, allows for gel formation by ionic cross-linking with divalent cations like calcium ions, generating a stable "egg-box" structure. The effects of pH, temperature, and ion concentration on gelation are explored, as well as other gel forms such as in situ and heat-sensitive gels. Alginate is widely used in the medical and pharmaceutical areas to promote tissue engineering through cell encapsulation and scaffolding, as well as in drug delivery systems for controlled and targeted release. In GERD therapy, alginate produces a gel raft that inhibits acid reflux, providing an effective alternative to proton pump inhibitors. Alginate-based products have demonstrated clinical success, strengthening alginate's medicinal promise. The review also discusses alginate-related issues, such as source variability and stability, as well as innovative modifications to improve treatment effects. These improvements establish alginate as a potential material for customized medication and tailored delivery systems.
Collapse
Affiliation(s)
- Devesh U Kapoor
- Dr. Dayaram Patel Pharmacy College, Bardoli, Gujarat 394601, India
| | - Anil Pareek
- Department of Pharmaceutics, Lachoo Memorial College of Science and Technology (Autonomous), Jodhpur, Rajasthan 342003, India
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, Rajasthan 304022, India
| | - Bhupendra G Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva, Gujarat 384012, India; Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura 140401 Punjab, India.
| | | | - Pornsak Sriamornsak
- Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand; Academy of Science, The Royal Society of Thailand, Bangkok 10300, Thailand; Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu 602105, India.
| |
Collapse
|
13
|
Verçosa BLA, Muniz-Junqueira MI, Borges LDF, Melo MN, Vasconcelos AC. Enhanced apoptosis, inflammatory cellularity, collagen deposition, and interaction between fibroblasts and Leishmania amastigotes in undamaged ear skin of dogs with leishmaniosis. Vet Parasitol 2025; 337:110488. [PMID: 40315687 DOI: 10.1016/j.vetpar.2025.110488] [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: 03/13/2025] [Revised: 04/25/2025] [Accepted: 04/27/2025] [Indexed: 05/04/2025]
Abstract
Fibroblasts are located close to the area of skin inoculation of Leishmania promastigotes. They are a potential cellular target for early parasite infection, harboring amastigotes of Leishmania spp. This study aimed to determine the apoptosis in fibroblasts, and to correlate these results with inflammation, parasite load, AgNOR (Argyrophilic Nucleolar Organizer Region) index, and clinical features in Leishmania-affected dogs. Fragments from the undamaged ear skin of 16 Leishmania-infected and seven uninfected dogs were evaluated by histomorphometry and immunohistochemical analysis, which correlated fibroblast apoptosis to clinical manifestation and parasite load. Ultra-thin sections were examined under transmission electronic microscopy (TEM). When applying immunohistochemical analysis, Leishmania amastigotes were only found in clinically affected dogs. The cellularity of the inflammatory infiltrate and the AgNOR index (fibroblasts and inflammatory infiltrate) were higher in clinically affected dogs. The collagen deposition score was statistically significantly higher in Leishmania-infected dogs. The apoptotic index of inflammatory cells and fibroblasts proved to be higher in clinically affected dogs. From an ultrastructural point of view, apoptotic cells shrank, while the nuclear chromatin and cytoplasm condensed. Amastigotes were observed within inflammatory cells (neutrophils and macrophages) and in the inner portions of fibroblasts. Fibroblast apoptosis was related to both the increase in the parasite load and the intensity of the inflammatory response. Histomorphometric assessments (inflammation, parasite load, AgNOR index, and apoptosis) and clinical manifestations were also associated. Collagen deposition was positively correlated with AgNOR expression and the apoptotic index (inflammatory cell and fibroblast). Therefore, fibroblast apoptosis contributes to the infection process, pathogenesis, and chronicity of canine leishmaniosis. Moreover, fibroblasts may well provide an escape mechanism for immune defenses against Leishmania.
Collapse
Affiliation(s)
- Barbara Laurice Araújo Verçosa
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil; Laboratório de Imunologia Celular, Faculdade de Medicina, Universidade de Brasília, Brasília, Brazil.
| | | | - Luciano de F Borges
- Departamento de Ciências Biológicas, Universidade Federal de São Paulo, Diadema, São Paulo, Brazil.
| | - Maria Norma Melo
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| | - Anilton Cesar Vasconcelos
- Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
14
|
Sharma A, Balde A, Nazeer RA. A review on animal venom-based matrix metalloproteinase modulators and their therapeutic implications. Int Immunopharmacol 2025; 157:114703. [PMID: 40300352 DOI: 10.1016/j.intimp.2025.114703] [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: 01/18/2025] [Revised: 04/03/2025] [Accepted: 04/19/2025] [Indexed: 05/01/2025]
Abstract
Matrix Metalloproteinases (MMPs) belong to a family of proteolytic enzymes that degrade extracellular matrix components, such as collagen, elastin, laminin, and fibronectin. They also play a part in tissue remodeling by cleaving and rejoining the tissue proteins. Cancer, neurodegenerative disorders, cardiovascular diseases, arthritis, and chronic inflammatory conditions are just some of the diseases that can start or get worse when different MMPs are not working properly. Venomous Animals such as honeybees, toads, snakes, spiders, scorpions, jellyfish, and sea anemones contain venom-secreting glands, which help them defend against predators and immobilize their prey. The molecules that come from animal venom are a complicated mix of bioactive molecules, such as peptides, enzymes, proteins, and small organic compounds that do a number of biological things. Venom-derived molecules have been found to modulate MMP. These venoms and their components target specific signaling pathways, modifying MMP expression levels to either induce inflammation or exhibit anti-inflammatory effects. In this review, we study and explore different MMPs, such as MMP1, MMP2, MMP3, MMP7, MMP8, and MMP9, and their roles in the progression of certain diseases. We also look at different types of molecules derived from marine and land animal venom that are used as MMP modulators. We look at how they work by targeting specific signaling pathways to change MMPs and how they might be used as a medicine to stop diseases by decreasing MMPs.
Collapse
Affiliation(s)
- Ansumaan Sharma
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Akshad Balde
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Rasool Abdul Nazeer
- Biopharmaceuticals Lab, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
15
|
Degu B, Tesfaye B, Abebe W, Assefa K. Diversity of Ethiopian Black Cumin ( Nigella sativa L.) Based on Compositions of Essential Oil. Biochem Res Int 2025; 2025:2065593. [PMID: 40330625 PMCID: PMC12052447 DOI: 10.1155/bri/2065593] [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: 02/26/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Black cumin (Nigella sativa L.) seeds essential oil compositions (EOCs) have been used for their medicinal and aromatic values across the world since ancient times. Studies have revealed the presence of variability among black cumin genotypes in EOCs. In Ethiopia, few studies have been conducted to explore the variability of black cumin genotypes by using EOCs. This study investigated the variability of Ethiopian black cumin genotypes (EBCGs) by EOCs. Seeds of 64 N. sativa genotypes were used for this experiment. Composite samples of 100 g of seeds were collected and roughly ground from each genotype. The extraction was made by hydrodistillation using a Clevenger-type apparatus for 3 h, and the essential oil was collected by measuring the amount using a measuring pipette. The essential oil samples were stored in a refrigerator at 4°C until gas chromatography-mass spectrometry (GC-MS) analysis. Descriptive statistics was used to estimate the variations among populations' combined mean values of EOCs using the SAS version 9.4 software package. The correlation and hierarchical clustering analysis were made based on the combined mean values of EOCs using the R-software version 4.2.2 packages. A total of 21 EOCs were detected from the essential oil of 64 EBCGs using GC-MS, out of which ρ-cymene, thymoquinone, α-thujene, carvacrol, trans-4-methoxythujane, longifolene, terpinen-4-ol, β-pinene, α-pinene, and d-limonene dominated the essential oils. Among these, ρ-cymene, thymoquinone, α-thujene, trans-4-methoxythujane, and carvacrol were the most abundant constituents in all genotypes, while the rest varied among the genotypes. It is predicted that the major EOCs will be improved by 25.33%-152.14% over improved varieties by selecting the top 5% of landraces. The abundant EOC thymoquinone had a significant and positive correlation with carvacrol and a strong and significant negative correlation with α-thujene, α-pinene, β-pinene, ρ-cymene, and d-limonene. Based on the major EOCs, cluster analysis grouped the 64 genotypes into two different chemotypes. Cluster-I: Chemotype A is characterized by a high content of thymoquinone. Cluster-II: Chemotype B is characterized by a high content of ρ-cymene. The presence of the most abundant volatile constituents in genotypes 242835, 9068, and 014_ATH means they are essential for the pharmaceutical and food industries. This study disclosed the existence of a significant diversity of EOCs among the Ethiopian N. sativa genotypes, which can be exploited for future improvement programs.
Collapse
Affiliation(s)
- Basazinew Degu
- Wondo Genet Agricultural Research Center, Ethiopian Institute of Agricultural Research, P.O. Box 198, Shashemene, Ethiopia
- School of Plant and Horticultural Science, Hawassa University, P.O. Box 05, Hawassa, Ethiopia
| | - Bizuayehu Tesfaye
- School of Plant and Horticultural Science, Hawassa University, P.O. Box 05, Hawassa, Ethiopia
| | - Wendawek Abebe
- Department of Molecular, Cellular, and Microbial Biology, Addis Ababa University, P.O. Box 345, Addis Ababa, Ethiopia
| | - Kebebew Assefa
- Debre Zeit Agricultural Research Centre, Ethiopian Institute of Agricultural Research, P.O. Box 32, Debre Zeit, Ethiopia
| |
Collapse
|
16
|
Marciniak M, Stachowicz-Suhs M, Wagner M. The role of innate immune cells in modulating vascular dynamics in skin malignancies. Biochim Biophys Acta Rev Cancer 2025; 1880:189331. [PMID: 40280501 DOI: 10.1016/j.bbcan.2025.189331] [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: 01/28/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
A developing tumor relies heavily on blood vessels to supply oxygen and nutrients. As a result, angiogenesis, the formation of new blood vessels, supports tumor growth and progression. Similarly, lymphangiogenesis, the formation of new lymphatic vessels, plays a critical role in metastatic dissemination by providing pathways for malignant cells to spread. The tumor microenvironment is crucial for establishing and maintaining these vascular networks, with innate immune cells playing a key regulatory role. Notably, immune cells are specifically enriched in barrier tissues, such as the skin, emphasizing their importance in skin malignancies. Therefore, understanding their role in regulating angiogenesis and lymphangiogenesis is essential for developing novel therapeutic strategies. This review article explores how innate immune cells influence tumor vasculature and highlights the therapeutic potential that may arise from this knowledge.
Collapse
Affiliation(s)
- Mateusz Marciniak
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland; Department of Biochemistry and Immunochemistry, Wrocław Medical University, Wrocław, Poland
| | - Martyna Stachowicz-Suhs
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland
| | - Marek Wagner
- Innate Immunity Research Group, Life Sciences and Biotechnology Center, Łukasiewicz Research Network - PORT Polish Center for Technology Development, Wrocław, Poland.
| |
Collapse
|
17
|
Shannon AE, Teodorescu RN, Song NJ, Heil LR, Jacob CC, Remes PM, Li Z, Rubinstein MP, Searle BC. Rapid assay development for low input targeted proteomics using a versatile linear ion trap. Nat Commun 2025; 16:3794. [PMID: 40263265 PMCID: PMC12015518 DOI: 10.1038/s41467-025-58757-8] [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: 07/08/2024] [Accepted: 04/02/2025] [Indexed: 04/24/2025] Open
Abstract
Advances in proteomics and mass spectrometry enable the study of limited cell populations, where high-mass accuracy instruments are typically required. While triple quadrupoles offer fast and sensitive low-mass specificity measurements, these instruments are effectively restricted to targeted proteomics. Linear ion traps (LITs) offer a versatile, cost-effective alternative capable of both targeted and global proteomics. Here, we describe a workflow using a hybrid quadrupole-LIT instrument that rapidly develops targeted proteomics assays from global data-independent acquisition (DIA) measurements without high-mass accuracy. Using an automated software approach for scheduling parallel reaction monitoring assays (PRM), we show consistent quantification across three orders of magnitude in a matched-matrix background. We demonstrate measuring low-level proteins such as transcription factors and cytokines with quantitative linearity below two orders of magnitude in a 1 ng background proteome without requiring stable isotope-labeled standards. From a 1 ng sample, we found clear consistency between proteins in subsets of CD4+ and CD8+ T cells measured using high dimensional flow cytometry and LIT-based proteomics. Based on these results, we believe hybrid quadrupole-LIT instruments represent a valuable solution to expanding mass spectrometry in a wide variety of laboratory settings.
Collapse
Affiliation(s)
- Ariana E Shannon
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
- Department of Biomedical Informatics, The Ohio State University Medical Center, Columbus, OH, 43210, USA
| | - Rachael N Teodorescu
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - No Joon Song
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | | | | | | | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
| | - Mark P Rubinstein
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Brian C Searle
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, 43210, USA.
- Department of Biomedical Informatics, The Ohio State University Medical Center, Columbus, OH, 43210, USA.
| |
Collapse
|
18
|
Hu Z, Li W, Wei L, Ma J. Lactoferrin in cancer: Focus on mechanisms and translational medicine. Biochim Biophys Acta Rev Cancer 2025; 1880:189330. [PMID: 40274081 DOI: 10.1016/j.bbcan.2025.189330] [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: 12/05/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Lactoferrin is an iron-binding glycoprotein that provides natural protective effects to the human body. Its biological properties, including antibacterial, antiviral, anti-inflammatory, immune-regulatory, and iron metabolism-regulating functions, have been extensively studied. With further research, lactoferrin's impact on tumorigenesis and tumor microenvironment has become increasingly evident, as it inhibits tumor proliferation, invasion, and metastasis through multiple pathways. This article summarizes the molecular mechanisms underlying lactoferrin's anticancer effects, explores its association with the malignant progression of various cancers, and highlights its clinical translational potential as a potential cancer biomarker and drug delivery carrier to enhance anticancer therapy efficiency. Due to the high safety profile of lactoferrin, its widespread application in the field of cancer treatment is highly anticipated.
Collapse
Affiliation(s)
- Zhengyu Hu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medicine Sciences, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Wenchao Li
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medicine Sciences, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Lingyu Wei
- Laboratory of Clinical Research Center, Department of Pathology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China.
| | - Jian Ma
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medicine Sciences, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China.
| |
Collapse
|
19
|
Li S, Zhang J, Wei W, Zhang Z, Huang W, Xia L. The important role of myeloid-derived suppressor cells: From hepatitis to liver cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189329. [PMID: 40262654 DOI: 10.1016/j.bbcan.2025.189329] [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: 09/26/2024] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 04/24/2025]
Abstract
Liver homeostasis is coordinated by crosstalk between resident and infiltrating inflammatory cells. Liver disease creates a dynamic inflammatory microenvironment characterized by aberrant metabolism and continuous hepatic regeneration, making it an important risk factor for hepatocellular carcinoma (HCC) as well as liver failure. Recent studies have revealed a critical heterogeneous population of myeloid-derived suppressor cells (MDSCs), which influence liver disease progression and malignancy by dynamically regulating the immune microenvironment. MDSCs play an important role in preventing excessive immune responses in the liver. However, MDSCs are also associated with the promotion of liver injury and liver cancer progression. The plasticity of MDSCs in liver disease is a unique challenge for therapeutic intervention strategies and requires a deeper understanding of the underlying mechanisms. Here, we review the role of MDSCs in the establishment and progression of liver disease and highlight the evidence for MDSCs as a priority target for current and future therapeutic strategies. We explore the fate of MDSCs from hepatitis to liver cancer, providing recent insights into potential targets for clinical intervention.
Collapse
Affiliation(s)
- Siwen Li
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Jiaqian Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Wang Wei
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
| | - Zhicheng Zhang
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| | - Wenjie Huang
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Clinical Medicine Research Center for Hepatic Surgery of Hubei Province, Key Laboratory of Organ Transplantation, Ministry of Education and Ministry of Public Health, Wuhan, Hubei 430030, China.
| | - Limin Xia
- Department of Gastroenterology, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China.
| |
Collapse
|
20
|
Lin G, Yao Z, Kang K, Luo R, Yi L, Lu Y. Dynamic evolution and antitumor mechanisms of CXCR6 +CD8 + T cells in small cell lung cancer treated with low-dose radiotherapy and immunotherapy. J Transl Med 2025; 23:453. [PMID: 40247265 PMCID: PMC12007177 DOI: 10.1186/s12967-025-06450-1] [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: 01/23/2025] [Accepted: 04/03/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Patients with small-cell lung cancer (SCLC) have the poor prognosis. Current research suggested that low-dose radiotherapy (LDRT) combined with immunotherapy can enhance the immunogenicity of tumor cells, thereby improving antigen presentation and promoting the intratumoral infiltration of CD8+ T cells, which significantly extends the survival of patients. However, the change trajectory of T cells, and the mechanisms underlying the promotion of intratumoral infiltration of CD8+ T cells, and the enhancement of their cytotoxic functions remain to be elucidated. METHODS To delineate the dynamic changes of T cells, we collected tumors from Kaede tumor-bearing mice that had undergone radioimmunotherapy. Using flow cytometry, we sorted intratumoral-infiltrating immune cells, which were required for single-cell RNA sequencing, at various time points (Kaede Red: derived from tumor-draining lymph node [TDLN]). The results obtained from the sequencing analysis were further validated through experiments, such as flow cytometry, immunofluorescence, and analysis of clinical cohort data. RESULTS Here, we observed stem-like T cells migrating from the TDLN to the tumor site and differentiating into effector phenotypes within the tumor. Dendritic cells (DCs) are the key cluster that induces the differentiation of stem-like T cell into effector phenotypes. Moreover, SCLC patients with a high infiltration of tumor-specific CXCR6+CD8+ T cells exhibited a supportive TME and longer survival time (P < 0.001). CONCLUSIONS This study delineates the change trajectory of CD8+ T cells, identifies the crucial role of DCs in T cell differentiation, and highlights the significance of tumor-specific CXCR6+CD8+ T cells in anti-tumor immunity. Future therapeutic strategies for SCLC could focus on enhancing the infiltration of activated DCs and CXCR6+CD8+ T cells within the tumor microenvironment to improve treatment efficacy.
Collapse
Affiliation(s)
- Guo Lin
- Division of Thoracic Tumor Multimodality Treatment and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhuoran Yao
- Division of Thoracic Tumor Multimodality Treatment and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Kai Kang
- Division of Thoracic Tumor Multimodality Treatment and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ren Luo
- Division of Thoracic Tumor Multimodality Treatment and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Linglu Yi
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment and Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| |
Collapse
|
21
|
Zhang Y, Li T, Yang C, Pan Q, Pan C, Zhang X, Zhang Y, Shu X, Wang Z, He Z, Qu Z, Song C. Fermented Erigeron breviscapus flavonoids: anti-pseudorabies virus efficacy and mechanisms in vitro and in vivo. Front Vet Sci 2025; 12:1562879. [PMID: 40308697 PMCID: PMC12042226 DOI: 10.3389/fvets.2025.1562879] [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: 02/04/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Introduction Erigeron breviscapus exhibits anti-inflammatory properties, protects neuronal cells and enhances immune function. Modern traditional Chinese medicine fermentation techniques can increase the bioactive compound content in Erigeron breviscapus. However, its potential therapeutic effects against the porcine pseudorabies virus (PRV) remain unclear. Methods A PRV infection model was established in mouse trigeminal ganglion (TG) cells to determine the optimal antiviral mode of action of flavonoids from fermented Erigeron breviscapus (FEBF). Additionally, a PRV-infected rat model was developed to evaluate the in vivo antiviral efficacy of FEBF. Results FEBF demonstrated a higher protective rate and a lower viral copy number compared to unfermented E. breviscapus flavonoids (EBF). The protective effect was most pronounced under toxicological and inhibitory conditions, surpassing the blocking effect. PRV infection upregulated TLR4, MyD88, and NF-κB p65 protein expression during the pre-infection phase, followed by their downregulation after 12 h. FEBF regulated PRV-induced changes in protein expression, restoring them to near-normal levels by 36 h. In vivo assessments of pathological injury, PRV viral load, neuronal count, and neuronal apoptosis indicated that FEBF provided superior neuroprotection compared to both Minocycline (MINO), a broad-spectrum neuroprotective drug, and unfermented EBF. Mechanistic studies further revealed that FEBF modulated microglial polarization and regulated the inflammatory cytokines IL-6, TNF-α, IL-4, and IL-10. Conclusion These findings demonstrate that FEBF exhibits significant antiviral effects against PRV in both in vitro and in vivo models. FEBF represents a promising candidate for the development of anti-PRV therapeutics.
Collapse
Affiliation(s)
- Ying Zhang
- College of Veterinary Medicine of Yunnan Agricultural University, Kunming, Yunnan, China
| | - Ting Li
- College of Veterinary Medicine of Yunnan Agricultural University, Kunming, Yunnan, China
| | - Chunkun Yang
- Yongshan County Animal Husbandry Technology Extension Station, Zhaotong, Yunnan, China
| | - Qiong Pan
- College of Veterinary Medicine of Yunnan Agricultural University, Kunming, Yunnan, China
| | - Changxu Pan
- College of Veterinary Medicine of Yunnan Agricultural University, Kunming, Yunnan, China
| | - Xue Zhang
- College of Animal Science and Technology of Yunnan Agricultural University, Kunming, Yunnan, China
| | - Ying Zhang
- College of Veterinary Medicine of Yunnan Agricultural University, Kunming, Yunnan, China
| | - Xianghua Shu
- College of Veterinary Medicine of Yunnan Agricultural University, Kunming, Yunnan, China
| | - Zheng Wang
- Diqing Tibetan Autonomous Prefecture Animal Husbandry and Veterinary Scientific Research Institute, Shangri-la, Yunnan, China
| | - Zhenghong He
- Shangri-la Agriculture and Rural Bureau Animal Husbandry and Aquatic Technology Promotion Station, Shangri-la, Yunnan, China
| | - Zichen Qu
- Shangri-la Agriculture and Rural Bureau Animal Husbandry and Aquatic Technology Promotion Station, Shangri-la, Yunnan, China
| | - Chunlian Song
- College of Veterinary Medicine of Yunnan Agricultural University, Kunming, Yunnan, China
| |
Collapse
|
22
|
Shu H, Liao Q, Chen Z, Liang M, Zhang S, Liu J, Wu Y, Hu P, Luo M, Zhu W, Zhu X, Yang L, Yan T. Flavonoids serve as a promising therapeutic agent for ischemic stroke. Brain Res 2025; 1853:149528. [PMID: 39999903 DOI: 10.1016/j.brainres.2025.149528] [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: 01/08/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 02/27/2025]
Abstract
Ischemic stroke (IS) continues to be a major public health concern and is characterized by significantly high mortality and disabling rates. Inhibiting nerve cells death and enhancing the repair of ischemic tissue are important treatment concepts for IS. Currently, the mainstream treatment strategies mainly focus on short-term care, which underscores the urgent need for novel therapeutic strategies for long-term care. Emerging data reveal that flavonoids have surfaced as promising candidates for IS patients' long-term care. Flavonoids can alleviate neuroinflammation and anti-apoptosis due to their characteristic pharmacological mechanisms. Clinical evidence suggests that long-term flavonoids intake improves IS patients' long-term outcomes. Though the effect of flavonoids in IS treatment has been explored for decades, the neuroprotective pharmacodynamics have not been well established. Thereby, the aim of current review is to summarize the pathways involved in neuroprotective effect of flavonoids. This review will also advance the potential of flavonoids as a viable clinical candidate for the treatment of IS.
Collapse
Affiliation(s)
- Hongxin Shu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Qiuye Liao
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zhihao Chen
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Mingyu Liang
- School of life sciences, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Si Zhang
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Junzhe Liu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yanze Wu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ping Hu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Ming Luo
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Wenping Zhu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xingen Zhu
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Li Yang
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Tengfeng Yan
- Department of Neurosurgery, the 2st affiliated hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330006, China; Jiangxi Key Laboratory of Neurological Tumors and Cerebrovascular Diseases, Nanchang, Jiangxi 330006, China; Jiangxi Health Commission Key Laboratory of Neurological Medicine, Nanchang, Jiangxi 330006, China; Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi 330006, China.
| |
Collapse
|
23
|
Sun Y, Fang W, Peng J, Liu X, Wang C, Song L, Deng Z. Potential role of CFLAR in enhancing 5-FU sensitivity and modulating immune cell infiltration in breast cancer. Eur J Med Res 2025; 30:265. [PMID: 40211399 PMCID: PMC11983979 DOI: 10.1186/s40001-025-02532-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Breast cancer (BRCA), the most common malignancy among women, is a highly heterogeneous disease. Chemoresistance is a major factor leading to treatment failure in BRCA. However, mechanisms underlying the development of chemoresistance remain unclear. METHODS In this study, we performed a comprehensive bioinformatic analysis to examine the role of cell death-associated genes in BRCA treatment. Specifically, we focused on caspase 8 and Fas-associated protein with death domain-like apoptosis regulator (CFLAR), which was identified as a co-differentially expressed cell death-associated molecule with potential prognostic values. We then validated these findings through in vitro experiments in BT- 549 and MDA-MB- 231 breast cancer cells. RESULTS Based on bioinformatics analysis, CFLAR expression was found to be downregulated in patients with BRCA, whereas its high expression was significantly associated with improved prognosis. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated that aberrantly expressed CFLAR was potentially associated with oxidative phosphorylation, T cell receptor signaling, and NADH dehydrogenase (ubiquinone) activity. In vitro experiments demonstrated that overexpression of CFLAR inhibited the generation of reactive oxygen species (ROS), consequently promoting 5-fluorouracil (5-FU) sensitivity in BT- 549 and MDA-MB- 231 breast cancer cells. The expression of CFLAR was positively correlated with the abundance of several tumor-infiltrating immune cells, especially CD8 + T cells, further supporting the role of CFLAR in immune regulation. CONCLUSION In conclusion, this study reveals the novel roles of CFLAR in enhancing chemotherapy sensitivity and patient outcome in BRCA and underscores its potential as a therapeutic target. These results supported CFLAR as a therapeutic target and prognostic biomarker in BRCA patients.
Collapse
Affiliation(s)
- Yuwei Sun
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Weilun Fang
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jinwu Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Xingling Liu
- Department of Pharmacy, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, Guangdong, China.
| | - Chunjiang Wang
- Department of Pharmacy, the Third Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Liying Song
- Department of Pharmacy, the Third Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhenzhen Deng
- Department of Pharmacy, the Third Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| |
Collapse
|
24
|
Che Y, Song B, Song Y, Sun J, Zhao X, Zheng J, Shi Y, Duan Y, Shao Y, Yang T, Yang L. Thymoquinone enhances efficacy of cervical Cancer therapeutic vaccines via modulating CD8 + T cells. Int Immunopharmacol 2025:114605. [PMID: 40316484 DOI: 10.1016/j.intimp.2025.114605] [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: 02/06/2025] [Revised: 03/15/2025] [Accepted: 03/29/2025] [Indexed: 05/04/2025]
Abstract
Cervical cancer, primarily driven by persistent high-risk human papillomavirus (HPV) infection, remains a global health challenge. While therapeutic vaccines offer promising alternatives to conventional treatments, their clinical efficacy is often hindered by limitations in antigen presentation and the immunosuppressive tumor microenvironment (TME). In this study, we explore the potential of thymoquinone (TQ), a bioactive compound derived from Nigella sativa seeds, to enhance the efficacy of cervical cancer therapeutic vaccines. Utilizing a mouse cervical cancer xenograft model with TC-1 cells expressing HPV16 E6/E7 and ras genes, we observed substantial heterogeneity in vaccine-induced antitumor responses. Mice were stratified into hyporesponsiveness (Hypo) and hyperresponsiveness (Hyper) groups based on tumor progression. The Hyper group exhibited significantly reduced tumor growth, marked by increased CD4+ and CD8+ T cell infiltration, reduced regulatory T cells (Tregs), and altered systemic immune parameters. Metabolite profiling identified 63 differential metabolites, with TQ being notably upregulated in the Hyper group, correlating positively with CD4+ and CD8+ T cells and negatively with Tregs and tumor volume. Combining TQ with the vaccine potentiated antitumor responses, leading to smaller tumors without significant changes in body weight. Immunohistochemical and flow cytometry analyses revealed augmented CD4+ and CD8+ T cell infiltration and reduced Tregs, alongside increased IFN-γ production in the combination of vaccine with (Vax + TQ) group. RNA-Seq analysis of TQ-treated CD8+ T cells revealed upregulation of immune-related genes, including Spp1, Grem1, Cxcl10, Isg15, Lcn2, and Sema3c, highlighting enriched immune pathways. Furthermore, TQ treatment upregulated ISG15 expression and enhanced CD8+ T cell cytotoxicity and cytokine secretion both in vitro and in vivo. In conclusion, our findings suggest that TQ enhances the efficacy of cervical cancer therapeutic vaccines by modulating immune responses, particularly by upregulating ISG15 and boosting CD8+ T cell function. This study provides a theoretical foundation for TQ as a promising adjuvant in cancer immunotherapy.
Collapse
Affiliation(s)
- Yuxin Che
- Department of Microbiology and Immunology, Shanxi Medical University, China; Higher Education Key Laboratory of Tumor Immunology & Targeted Drug Development in Shanxi Province, Shanxi Medical University, Taiyuan 030001, China
| | - Bing Song
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China
| | - Yikun Song
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China
| | - Jiaying Sun
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China
| | - Xueting Zhao
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China
| | - Jinxiu Zheng
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China
| | - Yu Shi
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China; Higher Education Key Laboratory of Tumor Immunology & Targeted Drug Development in Shanxi Province, Shanxi Medical University, Taiyuan 030001, China
| | - Yujing Duan
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China; Higher Education Key Laboratory of Tumor Immunology & Targeted Drug Development in Shanxi Province, Shanxi Medical University, Taiyuan 030001, China
| | - Ying Shao
- Higher Education Key Laboratory of Tumor Immunology & Targeted Drug Development in Shanxi Province, Shanxi Medical University, Taiyuan 030001, China; Department of Pathophysiology, Shanxi Medical University, Taiyuan 030001, China
| | - Tao Yang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, China; Higher Education Key Laboratory of Tumor Immunology & Targeted Drug Development in Shanxi Province, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China.
| | - Lijun Yang
- Higher Education Key Laboratory of Tumor Immunology & Targeted Drug Development in Shanxi Province, Shanxi Medical University, Taiyuan 030001, China; Key Laboratory of Coal Environmental Pathogenicity and Prevention, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China; Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
25
|
Magdy G, Elattar RH, Abdel Salam RA, Hadad GM, El-Deen AK. Unlocking the power of nanohybrids: A critical review on carbon nanomaterial-functionalized silver nanoparticles for advanced antimicrobial applications. Colloids Surf B Biointerfaces 2025; 252:114678. [PMID: 40209605 DOI: 10.1016/j.colsurfb.2025.114678] [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: 02/14/2025] [Revised: 03/25/2025] [Accepted: 04/01/2025] [Indexed: 04/12/2025]
Abstract
Over the last decades, nanotechnology has enabled the development of several inorganic nanoparticles with significant biocidal properties against diverse microorganisms. Silver nanoparticles (AgNPs) are among the most promising antimicrobial nanomaterials that have attracted substantial attention in various fields due to their low cost, low toxicity, biocompatibility, photo and chemical stability, easy preparation, high fluorescence, and tunability. Carbon nanomaterials (CNMs) are another appealing nanomaterial with antimicrobial qualities. While the antimicrobial efficacy of both AgNPs and CNMs is well-established, there is significant interest in the creation of CNMs/AgNPs hybrid materials for several applications due to their potential to exhibit synergistic bactericidal properties that surpass the yields of their components. This review represents a general overview of the different kinds, characterization techniques, synthesis processes, and antimicrobial activity of CNMs/AgNPs, along with an analysis of their benefits, drawbacks, and antimicrobial applications. Researchers and scientists interested in learning more about the potential of CNMs/AgNPs for advanced antimicrobial applications are likely to find this review to be a valuable resource.
Collapse
Affiliation(s)
- Galal Magdy
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33511, Egypt; Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Mansoura National University, Gamasa 7731168, Egypt.
| | - Rehab H Elattar
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.
| | - Randa A Abdel Salam
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Ghada M Hadad
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Asmaa Kamal El-Deen
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Department of Agro-Environmental Sciences, Faculty of Agriculture, Kyushu University, Fukuoka 819-0395, Japan.
| |
Collapse
|
26
|
Naeem N, Mughal EU. Comprehensive assessment of 3-benzyloxyflavones as β-glucosidase inhibitors: in vitro, in vivo, kinetic, SAR and computational studies. RSC Adv 2025; 15:10484-10500. [PMID: 40190633 PMCID: PMC11969444 DOI: 10.1039/d4ra08558b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/15/2025] [Indexed: 04/09/2025] Open
Abstract
In this study, a series of 3-benzyloxyflavone derivatives (1-10) was designed and, for the first time, evaluated for both in vitro and in vivo inhibitory activity against the β-glucosidase enzyme. The enzyme inhibitory potential of these derivatives was further assessed in an antihyperglycemic context using in vivo mechanism-based assays on p-nitrophenyl-β-d-glucopyranoside (PGLT) induced diabetic models. Additionally, structure-activity relationship (SAR) was employed to identify structural features crucial for activity. Molecular docking analyses revealed that both the potent compounds and co-crystallized ligands shared similar binding orientations within the active sites of β-glucosidase (PDB IDs: 3AJ7; 66K1). Molecular dynamics (MD) simulations validated the stability of the inhibitor-enzyme complexes under physiological conditions, while density functional theory (DFT) calculations helped elucidate electronic properties critical for activity. Drug-likeness analysis was also conducted to assess the pharmacokinetic potential of the derivatives. The results highlighted several derivatives with significant inhibitory activity, desirable pharmacokinetic profiles, and promising drug-like properties, making them potential candidates for therapeutic development. The target derivatives (1-10) demonstrated strong potential as lead compounds for developing new anti-diabetic agents with effective anti-hyperglycemic properties.
Collapse
Affiliation(s)
- Nafeesa Naeem
- Department of Chemistry, University of Gujrat Gujrat-50700 Pakistan
| | | |
Collapse
|
27
|
Goleij P, Tabari MAK, Khandan M, Poudineh M, Rezaee A, Sadreddini S, Sanaye PM, Khan H, Larsen DS, Daglia M. Genistein in focus: pharmacological effects and immune pathway modulation in cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3557-3571. [PMID: 39601821 DOI: 10.1007/s00210-024-03647-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Cancer is a significant global health concern, responsible for mortality and morbidity of individuals. It is characterized by uncontrolled cellular growth, tumor formation, and potential metastasis. The immune system is pivotal in recognizing and eliminating cancerous cells, with immune cells such as T cells, B cells, natural killer cells (NK), and dendritic cells playing critical roles. Dysregulation of immune responses can contribute to cancer progression. Phytochemicals, bioactive compounds derived from plants, have gained attention for their potential roles in cancer prevention and therapy due to their antioxidant, anti-inflammatory, and immunomodulatory properties. Genistein, an isoflavone found in soy products, is of particular interest. In this study, genistein's mechanisms of action at the molecular and cellular levels in cancer were demonstrated, highlighting its impact on T and B lymphocytes, NK cells and dendritic cells. Genistein's ability to influence cytokine production, reducing levels of inflammatory cytokines such as TNF-α, IL-6, and IL-1β, is emphasized. Genistein modulates inflammatory response pathways like Toll-like receptors (TLRs), NF-κB, chemokines, and MAPK, inhibiting tumor growth, promoting apoptosis, and reducing metastasis. It shows promise in overcoming chemoresistance, particularly in ovarian and neuroblastoma cancers, by inhibiting autophagy. Genistein also affects T-cell execution markers, including granzyme B, TNF-α, and FAS ligand in cancer by influencing key proteins involved in immune response and apoptosis. Clinical trials have investigated genistein's therapeutic potential, revealing its promise in enhancing the efficacy of traditional cancer treatments while mitigating associated toxicities. Genistein helps overcome chemoresistance in various cancers by inhibiting autophagy and promoting apoptosis. It also enhances immunotherapy by boosting immune responses and modifying antigens, but careful dosing is needed when combined with anti-PD-1 treatments to avoid reducing effectiveness.
Collapse
Affiliation(s)
- Pouya Goleij
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, 6715847141, Iran.
- Department of Genetics, Faculty of Biology, Sana Institute of Higher Education, Sari, 4816118761, Iran.
- PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research, Network (USERN), Tehran, Iran.
| | - Mohammad Amin Khazeei Tabari
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, 4815733971, Iran
- USERN Office, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohanna Khandan
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Mazandaran, 4815733971, Iran
- USERN Office, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohadeseh Poudineh
- Student Research Committee, School of Medicine, Zanjan University of Medical Sciences, Zanjan, 4513956184, Iran
| | - Aryan Rezaee
- Medical Doctor, School of Medicine, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Sarvin Sadreddini
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, 51656-87386, Iran
| | - Pantea Majma Sanaye
- School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, 4513956184, Iran
| | - Haroon Khan
- Department of Pharmacy, Faculty of Chemical and Life Sciences, Abdul Wali Khan University Mardan, Mardan, 23200, Pakistan.
- Department of Pharmacy, Korea University, Sejong, 20019, South Korea.
| | - Danaé S Larsen
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland, 1010, New Zealand
| | - Maria Daglia
- Department of Pharmacy, University of Naples "Federico II", Via D. Montesano 49, Naples, 80131, Italy
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang, 212013, China
| |
Collapse
|
28
|
Mohsen S, El-Gilany AH, Essam N. Effect of Ramadan fasting on changes in smoking and vaping behaviors and withdrawal symptoms severity: a cross-sectional study. J Addict Dis 2025; 43:168-178. [PMID: 38790079 DOI: 10.1080/10550887.2024.2354565] [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: 05/26/2024]
Abstract
BACKGROUND Ramadan fasting, an obligatory duty for adult Muslims, entails refraining from eating, drinking, smoking, and vaping from dawn until sunset. Throughout Ramadan, people accept major changes to daily routines. Consequently, Ramadan is associated with abstinence symptoms and changes in smoking or vaping behavior. AIM The study aims to assess the presence of withdrawal symptoms and its severity, as well as smoking or vaping behavior change and its determinants during Ramadan fasting. METHODS A cross-sectional study was conducted during Ramadan in 2023. Data were collected online using a Google form, including questionnaires assessing socio-demographic data, nicotine dependence using Fagerstrom Test for Nicotine Dependence, vaping and shisha smoking, presence and severity of physiological and psychological withdrawal symptoms and its coping strategies. Smoking and vaping behavioral change during Ramadan and its contributing factors were also evaluated. RESULTS A total of 251 participants (96% males), mean age ± standard deviation (33 ± 12.1) were surveyed. Withdrawal symptoms were reported among 70.5%, however, their severity was generally low both physiologically and psychologically. Education, nationality, smoking duration, nicotine dependence, and the amount of consumed E-liquid per day were the independent predictors of withdrawal symptoms presence. Attempts to quit smoking and vaping were reported by 54.6% of the participants; young age and higher educational levels were the factors associated with more quitting attempts. CONCLUSIONS The withdrawal symptoms severity linked to Ramadan fasting is minimal. Many smokers and vapers made attempts to quit during Ramadan. Ramadan presents a chance for smoking and vaping cessation with some behavioral support.
Collapse
Affiliation(s)
- Shorouk Mohsen
- Department of Public Health and Community Medicine, Faculty of Medicine, Mansoura University, Mansoura City, Egypt
| | - Abdel-Hady El-Gilany
- Department of Public Health and Community Medicine, Faculty of Medicine, Mansoura University, Mansoura City, Egypt
| | - Noha Essam
- Department of Public Health and Community Medicine, Faculty of Medicine, Mansoura University, Mansoura City, Egypt
| |
Collapse
|
29
|
Aftabi S, Barzegar Behrooz A, Cordani M, Rahiman N, Sadeghdoust M, Aligolighasemabadi F, Pistorius S, Alavizadeh SH, Taefehshokr N, Ghavami S. Therapeutic targeting of TGF-β in lung cancer. FEBS J 2025; 292:1520-1557. [PMID: 39083441 PMCID: PMC11970718 DOI: 10.1111/febs.17234] [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: 11/05/2023] [Revised: 05/22/2024] [Accepted: 07/19/2024] [Indexed: 08/02/2024]
Abstract
Transforming growth factor-β (TGF-β) plays a complex role in lung cancer pathophysiology, initially acting as a tumor suppressor by inhibiting early-stage tumor growth. However, its role evolves in the advanced stages of the disease, where it contributes to tumor progression not by directly promoting cell proliferation but by enhancing epithelial-mesenchymal transition (EMT) and creating a conducive tumor microenvironment. While EMT is typically associated with enhanced migratory and invasive capabilities rather than proliferation per se, TGF-β's influence on this process facilitates the complex dynamics of tumor metastasis. Additionally, TGF-β impacts the tumor microenvironment by interacting with immune cells, a process influenced by genetic and epigenetic changes within tumor cells. This interaction highlights its role in immune evasion and chemoresistance, further complicating lung cancer therapy. This review provides a critical overview of recent findings on TGF-β's involvement in lung cancer, its contribution to chemoresistance, and its modulation of the immune response. Despite the considerable challenges encountered in clinical trials and the development of new treatments targeting the TGF-β pathway, this review highlights the necessity for continued, in-depth investigation into the roles of TGF-β. A deeper comprehension of these roles may lead to novel, targeted therapies for lung cancer. Despite the intricate behavior of TGF-β signaling in tumors and previous challenges, further research could yield innovative treatment strategies.
Collapse
Affiliation(s)
- Sajjad Aftabi
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Paul Albrechtsen Research Institute, CancerCare ManitobaUniversity of ManitobaWinnipegCanada
- Department of Physics and AstronomyUniversity of ManitobaWinnipegCanada
| | - Amir Barzegar Behrooz
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Electrophysiology Research Center, Neuroscience InstituteTehran University of Medical SciencesIran
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of BiologyComplutense UniversityMadridSpain
- Instituto de Investigaciones Sanitarias San Carlos (IdISSC)MadridSpain
| | - Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesIran
- Department of Pharmaceutical Nanotechnology, School of PharmacyMashhad University of Medical SciencesIran
| | - Mohammadamin Sadeghdoust
- Division of BioMedical Sciences, Faculty of MedicineMemorial University of NewfoundlandSt. John'sCanada
| | - Farnaz Aligolighasemabadi
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
| | - Stephen Pistorius
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Paul Albrechtsen Research Institute, CancerCare ManitobaUniversity of ManitobaWinnipegCanada
- Department of Physics and AstronomyUniversity of ManitobaWinnipegCanada
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology InstituteMashhad University of Medical SciencesIran
- Department of Pharmaceutical Nanotechnology, School of PharmacyMashhad University of Medical SciencesIran
| | - Nima Taefehshokr
- Apoptosis Research CentreChildren's Hospital of Eastern Ontario Research InstituteOttawaCanada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell ScienceUniversity of Manitoba College of MedicineWinnipegCanada
- Paul Albrechtsen Research Institute, CancerCare ManitobaUniversity of ManitobaWinnipegCanada
- Faculty Academy of Silesia, Faculty of MedicineKatowicePoland
- Children Hospital Research Institute of ManitobaUniversity of ManitobaWinnipegCanada
| |
Collapse
|
30
|
Liang Y, Zhao Y, Qi Z, Li X, Zhao Y. Ferroptosis: CD8 +T cells' blade to destroy tumor cells or poison for self-destruction. Cell Death Discov 2025; 11:128. [PMID: 40169575 PMCID: PMC11962101 DOI: 10.1038/s41420-025-02415-x] [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: 11/03/2024] [Revised: 02/19/2025] [Accepted: 03/19/2025] [Indexed: 04/03/2025] Open
Abstract
Ferroptosis represents an emerging, iron-dependent form of cell death driven by lipid peroxidation. In recent years, it has garnered significant attention in the realm of cancer immunotherapy, particularly in studies involving immune checkpoint inhibitors. This form of cell death not only enhances our comprehension of the tumor microenvironment but is also considered a promising therapeutic strategy to address tumor resistance, investigate immune activation mechanisms, and facilitate the development of cancer vaccines. The combination of immunotherapy with ferroptosis provides innovative targets and fresh perspectives for advancing cancer treatment. Nevertheless, tumor cells appear to possess a wider array of ferroptosis evasion strategies compared to CD8+T cells, which have been conclusively shown to be more vulnerable to ferroptosis. Furthermore, ferroptosis in the TME can create a favorable environment for tumor survival and invasion. Under this premise, both inducing tumor cell ferroptosis and inhibiting T cell ferroptosis will impact antitumor immunity to some extent, and even make the final result run counter to our therapeutic purpose. This paper systematically elucidates the dual-edged sword role of ferroptosis in the antitumor process of T cells, briefly outlining the complexity of ferroptosis within the TME. It explores potential side effects associated with ferroptosis-inducing therapies and critically considers the combined application of ferroptosis-based therapies with ICIs. Furthermore, it highlights the current challenges faced by this combined therapeutic approach and points out future directions for development.
Collapse
Affiliation(s)
- Yuan Liang
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yixin Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhaoyang Qi
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinru Li
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Yuguang Zhao
- Cancer Center, the First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
31
|
Moon CY, Belabed M, Park MD, Mattiuz R, Puleston D, Merad M. Dendritic cell maturation in cancer. Nat Rev Cancer 2025; 25:225-248. [PMID: 39920276 PMCID: PMC11954679 DOI: 10.1038/s41568-024-00787-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 02/09/2025]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells that are present at low abundance in the circulation and tissues; they serve as crucial immune sentinels by continually sampling their environment, migrating to secondary lymphoid organs and shaping adaptive immune responses through antigen presentation. Owing to their ability to orchestrate tolerogenic or immunogenic responses to a specific antigen, DCs have a pivotal role in antitumour immunity and the response to immune checkpoint blockade and other immunotherapeutic approaches. The multifaceted functions of DCs are acquired through a complex, multistage process called maturation. Although the role of inflammatory triggers in driving DC maturation was established decades ago, less is known about DC maturation in non-inflammatory contexts, such as during homeostasis and in cancer. The advent of single-cell technologies has enabled an unbiased, high-dimensional characterization of various DC states, including mature DCs. This approach has clarified the molecular programmes associated with DC maturation and also revealed how cancers exploit these pathways to subvert immune surveillance. In this Review, we discuss the mechanisms by which cancer disrupts DC maturation and highlight emerging therapeutic opportunities to modulate DC states. These insights could inform the development of DC-centric immunotherapies, expanding the arsenal of strategies to enhance antitumour immunity.
Collapse
Affiliation(s)
- Chang Yoon Moon
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meriem Belabed
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthew D Park
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raphaël Mattiuz
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Puleston
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
32
|
Parvanian S, Ge X, Garris CS. Recent developments in myeloid immune modulation in cancer therapy. Trends Cancer 2025; 11:365-375. [PMID: 39794212 DOI: 10.1016/j.trecan.2024.12.003] [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: 10/15/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/13/2025]
Abstract
Myeloid cells play a crucial dual role in cancer progression and response to therapy, promoting tumor growth, enabling immune suppression, and contributing to metastatic spread. The ability of these cells to modulate the immune system has made them attractive targets for therapeutic strategies aimed at shifting their function from tumor promotion to fostering antitumor immunity. Therapeutic approaches targeting myeloid cells focus on modifying their numbers, genetics, metabolism, and interactions within the tumor microenvironment. These strategies aim to reverse their suppressive functions and redirect them to support antitumor immune responses by inhibiting immunosuppressive pathways, targeting specific receptors, and promoting their differentiation into less immunosuppressive phenotypes. Here, we discuss recent approaches to clinically target tumor myeloid cells, focusing on reprogramming myeloid cells to promote antitumor immunity.
Collapse
Affiliation(s)
- Sepideh Parvanian
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114, USA
| | - Xinying Ge
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114, USA; Master's Program in Immunology Harvard Medical School, 200 Longwood Ave, Boston, MA 02115, USA
| | - Christopher S Garris
- Center for Systems Biology, Massachusetts General Hospital, 185 Cambridge St, CPZN 5206, Boston, MA 02114, USA; Department of Pathology, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114, USA.
| |
Collapse
|
33
|
Shabestari SM, Jafari SH, Benisi SZ, Khoeini R, Shojaei S, Ghorbani M, Goodarzi V. Role of phosphate-modified cellulose into the scaffold based on poly (glycerol azelaic acid)-co-poly(ε-caprolactone) for using bone regenerative medicine. Int J Biol Macromol 2025; 304:140855. [PMID: 39933675 DOI: 10.1016/j.ijbiomac.2025.140855] [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: 12/18/2024] [Revised: 01/19/2025] [Accepted: 02/08/2025] [Indexed: 02/13/2025]
Abstract
Advancements in bone tissue engineering critically depend on the development of specialized scaffolds that promote effective bone regeneration. This study introduces innovative scaffolds composed of poly (glycerol azelaic acid) (PGAz) and poly (caprolactone) (PCL), enhanced with varying concentrations of phosphate modified cellulose (PMC) nanofibers. Utilizing citric acid as a green cross-linker enhances the eco-friendliness and biocompatibility of these scaffolds. The molecular structures were verified using Hydrogen 1H NMR and Carbon 13CNMR spectroscopy. Fourier Transform Infrared (FTIR) Spectroscopy confirmed successful integration of vital phosphate groups for biological functionality. Morphological and compositional analysis through Field Emission Scanning Electron Microscopy (FESEM) and Energy-Dispersive X-ray (EDX) mapping revealed homogeneous phosphate distribution and increased porosity across the scaffolds. Thermogravimetric Analysis (TGA) demonstrated that higher PMC concentrations improve thermal stability, enhancing scaffold resilience. Mechanical testing showed that scaffolds with 5 % PMC provide an optimal balance of compressive strength, compressive modulus, and compressive strain, suitable for bone tissue engineering. Dynamic water contact angle studies highlighted significant hydrophilic enhancements, crucial for cell attachment and proliferation. Hydrolytic degradation tests indicated that increased PMC accelerates degradation rates, aligning with tissue regeneration timelines. Hematoxylin and Eosin (H&E) staining and MTT assays, along with FESEM imaging of L929 mouse fibroblast cells cultured on the scaffolds, confirmed biocompatibility and cellular proliferation, particularly with 5 % PMC. These findings underscore the potential of PGAz-co-PCL scaffolds in advancing bone tissue regeneration through superior mechanical, thermal, and biological properties.
Collapse
Affiliation(s)
- Salar Mohammadi Shabestari
- Department of Polymer, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Seyed Hassan Jafari
- Department of Polymer, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Soheila Zamanlui Benisi
- Department of Biomedical Engineering, Islamic Azad University, Central Tehran Branch, Tehran, Iran; Stem cells Research Center, Tissue Engineering and Regenerative Medicine Institute, Islamic Azad University, Central Tehran Branch, Tehran, Iran
| | - Romina Khoeini
- Department of Biomedical Engineering, Islamic Azad University, Central Tehran Branch, Tehran, Iran
| | - Shahrohk Shojaei
- Department of Biomedical Engineering, Islamic Azad University, Central Tehran Branch, Tehran, Iran
| | - Masoud Ghorbani
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Vahabodin Goodarzi
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
34
|
Bhatt J, Ghigo A, Hirsch E. PI3K/Akt in IPF: untangling fibrosis and charting therapies. Front Immunol 2025; 16:1549277. [PMID: 40248697 PMCID: PMC12004373 DOI: 10.3389/fimmu.2025.1549277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/13/2025] [Indexed: 04/19/2025] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a chronic, progressive lung disease characterized by abnormal epithelial repair, persistent inflammation, and excessive extracellular matrix deposition, leading to irreversible scarring and respiratory failure. Central to its pathogenesis is the dysregulation of the PI3K/Akt signaling pathway, which drives fibroblast activation, epithelial-mesenchymal transition, apoptosis resistance, and cellular senescence. Senescent cells contribute to fibrosis through the secretion of pro-inflammatory and profibrotic factors in the senescence-associated secretory phenotype (SASP). Current antifibrotic therapies, Nintedanib and Pirfenidone, only slow disease progression and are limited by side effects, highlighting the need for novel treatments. This review focuses on the role of PI3K/Akt signaling in IPF pathogenesis, its intersection with inflammation and fibrosis, and emerging therapeutic approaches targeting molecules along this pathway.
Collapse
Affiliation(s)
- Janki Bhatt
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
- Kither Biotech S.r.l., Turin, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
- Kither Biotech S.r.l., Turin, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center “Guido Tarone”, University of Turin, Turin, Italy
- Kither Biotech S.r.l., Turin, Italy
| |
Collapse
|
35
|
Lai YG, Liao HT, Chen YH, Huang SW, Liou YH, Wu ZQ, Liao NS. cGAS and STING in Host Myeloid Cells Are Essential for Effective Cyclophosphamide Treatment of Advanced Breast Cancer. Cancers (Basel) 2025; 17:1130. [PMID: 40227734 PMCID: PMC11987962 DOI: 10.3390/cancers17071130] [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: 02/13/2025] [Revised: 03/21/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cyclophosphamide (CTX) treatment in vivo kills proliferating tumor cells by DNA crosslinking; however, the suppression of tumor growth by CTX in several murine models requires CD8+ T cells. Given that CTX induces DNA damage and type I interferon (IFN-I), we investigated the role of host cGAS and STING in the anti-tumor effect of CTX in vivo. METHODS A metastasized EO771 breast cancer model with chromosomal instability and bone marrow (BM) chimera approach were used in this study. RESULTS We found that CTX therapy induces long-term survival of the mice, with this outcome being dependent on CD8+ T cells and cGAS/STING of BM-derived cells. Furthermore, the STING of type 1 conventional dendritic cells (cDC1s) and LysM+ cells and the IFN-I response of non-cDC1 myeloid cells are essential for CTX efficacy. We also found that the cGAS and STING of BM-derived cells positively modulate intratumoral exhausted and stem-cell-like CD8+ T cell populations under CTX treatment, with the latter only being affected by cGAS. CONCLUSIONS Our study demonstrates that the CD8+-T-cell-dependent anti-tumor mechanisms of CTX critically involve the cGAS-STING-IFN-I axis, IFN-I response, and STING-independent cGAS function in host myeloid cells. These findings suggest the deployment of CTX in treating advanced solid tumor to bypass the often-failed IFN-I production by tumor cells due to the chronic activation of intrinsic cGAS-STING caused by chromosomal instability.
Collapse
Affiliation(s)
- Yein-Gei Lai
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Hao-Ting Liao
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
- Department of Life Sciences, National Central University, Taoyuan 320, Taiwan
| | - Yung-Hsiang Chen
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Shih-Wen Huang
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Yae-Huei Liou
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Zhen-Qi Wu
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| | - Nan-Shih Liao
- Institute of Molecular Biology, Academia Sinica, Taipei 115, Taiwan; (Y.-G.L.); (H.-T.L.); (Y.-H.C.); (S.-W.H.); (Y.-H.L.); (Z.-Q.W.)
| |
Collapse
|
36
|
Hu E, Cheng R, Liu A, Wang Y, Long H, Hou J, Wang D, Wu W, Wu X. Metabolomic Profiles and Differential Constituents of Andrographis paniculata (Burm. f.) in Different Growth Stages and Parts. Molecules 2025; 30:1490. [PMID: 40286081 PMCID: PMC11990188 DOI: 10.3390/molecules30071490] [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: 02/18/2025] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025] Open
Abstract
Andrographis paniculata (Burm. f.) and its products have a long history of medicinal use in Asia. A. paniculata products are mainly made from the root extraction of stems, leaves and parts, but there may be differences in the proportion of different parts and different harvest times, which ultimately leads to certain differences in product quality. In this study, the chemical components and non-targeted metabolomics were characterized, and the characteristic compounds in different parts of A. paniculata at various growth stages were analyzed. By utilizing polygonal mass defect filtering, precursor ion lists, and a self-built compound library, a total of 225 components were identified in A. paniculata. Notably, spermidine derivatives and phosphatidylcholines were reported for the first time in this plant species. In total, 41 differential components were identified in different parts of A. paniculata. These findings provide scientific evidence for the selection of quality markers in A. paniculata and its products.
Collapse
Affiliation(s)
- Enming Hu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guiyang 550014, China; (E.H.); (D.W.)
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China; (R.C.); (A.L.); (Y.W.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Rui Cheng
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China; (R.C.); (A.L.); (Y.W.)
| | - Annian Liu
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China; (R.C.); (A.L.); (Y.W.)
| | - Ya Wang
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China; (R.C.); (A.L.); (Y.W.)
| | - Huali Long
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (H.L.); (J.H.)
| | - Jinjun Hou
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (H.L.); (J.H.)
| | - Daoping Wang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guiyang 550014, China; (E.H.); (D.W.)
- Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Wanying Wu
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (H.L.); (J.H.)
| | - Xingdong Wu
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Guiyang 550014, China; (E.H.); (D.W.)
- Guizhou Engineering Research Center of Industrial Key-Technology for Dendrobium Nobile, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China; (R.C.); (A.L.); (Y.W.)
- National Engineering Research Center of TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; (H.L.); (J.H.)
| |
Collapse
|
37
|
Yang J, Zhang H, Ni L, He J. Thymoquinone alleviates the accumulation of ROS and pyroptosis and promotes perforator skin flap survival through SIRT1/NF-κB pathway. Front Pharmacol 2025; 16:1567762. [PMID: 40201684 PMCID: PMC11975933 DOI: 10.3389/fphar.2025.1567762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
Perforator flap transplantation is an important technique in flap reconstructive surgery, but flap necrosis limits its clinical effectiveness. Thymoquinone (TQ), a natural bioactive plant quinone found in black seed, exhibits anti-inflammatory, angiogenic, and antimicrobial properties. This study investigates the therapeutic effects of TQ in a perforator flap model through in vivo and in vitro experiments. Human umbilical vein endothelial cells (HUVECs) were treated with Tert-butyl Hydroperoxide (TBHP) to simulate an in vitro flap model and were then treated with TQ. In vivo experiments used a rat perforator flap model, and vascularization was assessed using Doppler ultrasound on days 3 and 7 after flap creation. On day 7 post-surgery, flap samples were collected to evaluate vascularity, reactive oxygen species, apoptosis and pyroptosis. Network pharmacology analysis was conducted to identify relevant signaling pathways, and molecular docking techniques were used to predict potential target binding sites. In vitro results showed that both TQ treatment and NLRP3 inhibitors reduced the expression of pyroptosis-related proteins. In vivo results indicated that the TQ-treated group had increased flap survival area, blood flow intensity, and microvascular density, while oxidative stress, apoptosis, and pyroptosis levels were reduced. Angiogenesis was enhanced, and expression of the SIRT1 protein was increased, while the p-P65/NF-κB/NLRP3 pathway was downregulated. After treatment with a SIRT1 inhibitor, flap survival rate and angiogenesis were reduced. These findings suggest that TQ improves perforator flap survival by inhibiting the NF-κB/NLRP3 pathway and promoting angiogenesis.
Collapse
Affiliation(s)
- Jianxin Yang
- Department of Orthopaedics, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Haojie Zhang
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Libin Ni
- Department of Orthopaedics, Zhejiang Hospital, Hangzhou, Zhejiang, China
| | - Jun He
- Department of Orthopaedics, Zhejiang Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
38
|
Li T, Li T, Liang Y, Yuan Y, Liu Y, Yao Y, Lei X. Colorectal cancer cells-derived exosomal miR-188-3p promotes liver metastasis by creating a pre-metastatic niche via activation of hepatic stellate cells. J Transl Med 2025; 23:369. [PMID: 40134019 PMCID: PMC11938777 DOI: 10.1186/s12967-025-06334-4] [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: 12/18/2024] [Accepted: 03/01/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND/AIM Metastasis is the leading cause of mortality for colorectal cancer (CRC). Cancer-derived exosomes are widely recognized as the primary catalysts behind the development of pre-metastasis niche (PMN) in distal sites. However, the exact mechanism behind this process in CRC remains elusive. This study aimed to investigate the function and mechanisms underlying the role of exosomal miR-188-3p in activating hepatic stellate cells (HSCs) to develop the PMN and promote liver metastasis. METHODS We extracted exosomes from CRC cells using ultracentrifugation. Exosomes were identified using transmission electron microscopy, nanoparticle tracking analysis, and Western blot. Exosome uptake was assessed using fluorescence tracing, exosome PKH67 staining, and real-time quantitative PCR. The effects of CRC cell-derived exosomes on HSCs migration were evaluated using Transwell migration and wound healing assays. Key differentially expressed miRNAs were screened from the GEO database, and bioinformatics prediction along with dual-luciferase reporter assays were used to identify downstream target genes of miR-188-3p. Downstream related proteins of the target genes were detected by Western blot. In vivo, the distribution of exosomes and activation of HSCs in the liver were explored by tail vein injection of exosomes into nude mice. Further, the impact of exosomal miR-188-3p on liver metastasis was investigated using a spleen injection liver metastasis model. Finally, the expression levels of miR-188-3p in exosomes from CRC patient plasma were determined by real-time quantitative PCR, and the relationship between the expression of miR-188-3p in plasma exosomes and CRC prognosis was analyzed. RESULTS The expression level of miR-188-3p within plasma exosomes demonstrated a statistically significant increase in CRC with liver metastasis compared to those without liver metastases. We also demonstrated the transferability of miR-188-3p from CRC cells to HSCs cells via the exosomes. Exosomal miR-188-3p plays a pivotal role in orchestrating the establishment of PMN through targeting PHLPP2 to activate HSCs before tumor metastasis. Exosomal miR-188-3p was found to actively foster in vivo metastasis of CRC. Additionally, plasma exosomal miR-188-3p potentially serves as a viable blood-based biomarker for CRLM. CONCLUSION Exosomal miR-188-3p derived from CRC cells can promote liver metastasis by activating HSCs to form a PMN through targeting PHLPP2 to activate the AKT/mTOR pathway. These results offer a new perspective on the mechanisms driving CRLM.
Collapse
Affiliation(s)
- Tao Li
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Taiyuan Li
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yahang Liang
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yuli Yuan
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yang Liu
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yao Yao
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xiong Lei
- Department of General surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China.
- Gastrointestinal Surgical Institute, Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
39
|
Naeem N, Mughal EU. Pharmacological evaluation of 3-benzyloxyflavones for β-glucosidase inhibition: Experimental, kinetic and computational approaches. Int J Biol Macromol 2025:142317. [PMID: 40139594 DOI: 10.1016/j.ijbiomac.2025.142317] [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: 12/30/2024] [Revised: 01/29/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
β-Glucosidase is a crucial enzyme involved in carbohydrate metabolism, playing a key role in the hydrolysis of glycosidic bonds in dietary polysaccharides. Inhibition of β-glucosidase has emerged as a promising therapeutic strategy for managing postprandial hyperglycemia in diabetes by delaying/slowing glucose absorption and moderating blood sugar levels. In this study, a series of 3-benzyloxyflavone derivatives (1-10) was designed and, for the first time, evaluated for both in vitro and in vivo inhibitory activity against the β-glucosidase enzyme. The enzyme inhibitory potential of these derivatives was further assessed in an antihyperglycemic context using in vivo mechanism-based assays on p-nitrophenyl-β-D-glucopyranoside (PGLT) induced diabetic models. Additionally, structure-activity relationship (SAR) was employed to identify structural features crucial for activity. Molecular docking analyses revealed that both the potent compounds and co-crystallized ligands shared similar binding orientations within the active sites of β-glucosidase (PDB IDs: 3AJ7; 66K1). Molecular dynamics (MD) simulations validated the stability of the inhibitor-enzyme complexes under physiological conditions. Drug-likeness analysis was also conducted to assess the pharmacokinetic potential of the derivatives. We have also conducted Density Functional Theory (DFT) studies on the lead compounds to gain deeper insights into their electronic properties, structural stability, and interaction mechanisms with the target enzyme. The results highlighted several derivatives with significant inhibitory activity, desirable pharmacokinetic profiles, and promising drug-like properties, making them potential candidates for therapeutic development. The target derivatives (1-10) demonstrated strong potential as lead compounds for developing new anti-diabetic agents with effective anti-hyperglycemic properties.
Collapse
Affiliation(s)
- Nafeesa Naeem
- Department of Chemistry, University of Gujrat, Gujrat 50700, Pakistan
| | | |
Collapse
|
40
|
Kameni MN, Tchoupe EB, Kamdem SD, Bhalla N, Assam Assam JP, Tepa AN, Neba FR, Nanda RK, Awuah AAA, Amuasi JH, Netongo PM. Mutations in ace2 gene modulate cytokine levels and alter immune responses in Mycobacterium tuberculosis and SARS-CoV-2 co-infection: a Cameroonian cohort. Front Immunol 2025; 16:1533213. [PMID: 40196114 PMCID: PMC11973369 DOI: 10.3389/fimmu.2025.1533213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 02/27/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction SARS-CoV-2 and Mycobacterium tuberculosis (Mtb) share similarities in their modes of transmission, pathophysiological symptoms, and clinical manifestations. An imbalance in the immune response characterised by elevated levels of some inflammatory cytokines caused by tuberculosis (TB) and COVID-19 may increase the risk of developing a severe disease-like condition. It has been reported that TB increases the expression levels of Ace2 (angiotensin converting enzyme 2) and Tmprss2 (transmembrane protease serine 2) proteins, which are essential for COVID-19 pathogenesis. Single nucleotide polymorphisms (SNPs) variants of ace2 and tmprss2 genes can impact virus and host-cell interactions and alter immune responses by modulating cytokine production. This may modify the susceptibility and/or severity in COVID-19-infected people. The role of SNPs in ace2 and tmprss2 in relation to Mtb and SARS-CoV-2 co-infection is relatively underexplored. Method In this study, genotype frequency of 10 SNPs of ace2 and 03 SNPs of tmprss2 genes in a Cameroonian cohort consisting of COVID-19-positive (n = 31), TB-positive (n = 43), TB-COVID-19 co-infected (n = 21), and a control group (n = 24) were studied. The immune response was estimated by quantitating inflammatory cytokine levels alongside self-reported and clinically diagnosed symptoms. The relationship between specific genetic mutations in these ace2 gene SNPs and their impact on cytokine expression levels in Mtb and SARS-CoV-2 co-infected patients was investigated. Results We identified wild-type, heterozygous, and double-mutant genotypes in seven SNPs (rs2285666, rs6632677, rs4646116, rs4646140, rs147311723, rs2074192 and rs4646142) in ace2 gene, which showed significant variations in distribution across the study groups. Our most significant findings include the association of double mutant alleles (AA) of rs4646140 and rs2074192 in the ace2 gene with decreased IL-6 and IL-2 expression levels respectively in TB-COVID-19 participants. Also, the double mutant alleles (AA) of rs4646116 were responsible for increased expression level of IL-2 in TB-COVID-19 patients. Additionally, elevated serum levels of AST, urea, and D-dimer, as well as increased plasma concentrations of IL-10, IFN-γ, and TNF-α, have been associated with co-infections involving Mtb and SARS-CoV-2. Conclusion These biomarkers may reflect the complex interplay between the two pathogens and their impact on host immune responses and disease progression. This study highlights the critical role of genetic and immunological factors in shaping altered immune responses during co-infections involving Mtb and SARS-CoV-2. By elucidating these factors, the findings provide a foundation for a deeper understanding of host-pathogen interactions and their implications for disease progression and outcomes. Furthermore, this research has the potential to drive advancements in diagnostic approaches enabling more accurate detection and monitoring of co-infections.
Collapse
Affiliation(s)
- Mary Ngongang Kameni
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
- Department of Microbiology, University of Yaounde I, Yaounde, Cameroon
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Eric Berenger Tchoupe
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
- Department of Clinical Biochemistry, Faculty of Medicine and Biomedical Science, University of Yaounde I, Yaounde, Cameroon
| | - Severin Donald Kamdem
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
- Department of Pathology, School of Medicine, University of Utah, Salt Lake City, UT, United States
| | - Nikhil Bhalla
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | | | - Arnaud Njuiget Tepa
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
| | - Fuh Roger Neba
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
| | - Ranjan Kumar Nanda
- Translational Health Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Anthony Afum-Adjei Awuah
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - John Humphrey Amuasi
- Kumasi Centre for Collaborative Research in Tropical Medicine, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
- Department of Infectious Diseases Epidemiology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- College of Health Sciences, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Palmer Masumbe Netongo
- Molecular Diagnostics Research Group, Biotechnology Centre-University of Yaounde I (MDRG-BTC-UYI), Yaounde, Cameroon
- Biology Program, School of Science, Navajo Technical University, Crownpoint, NM, United States
- Department of Biochemistry, University of Yaounde I, Yaounde, Cameroon
| |
Collapse
|
41
|
Sornsenee P, Kooltheat N, Wongprot D, Suksabay P, Nam TG, Permpoon U, Saengsuwan P, Romyasamit C. Antibacterial, Antioxidant, and Anti-inflammatory Activities of Lacticaseibacillus paracasei Lysates Isolated from Fermented Palm Sap. Probiotics Antimicrob Proteins 2025:10.1007/s12602-025-10521-6. [PMID: 40120070 DOI: 10.1007/s12602-025-10521-6] [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] [Accepted: 03/10/2025] [Indexed: 03/25/2025]
Abstract
Paraprobiotics are inactivated microbial cells or cell fractions that confer health benefits to the consumer, and possess the ability to regulate both the adaptive and innate immune systems. They exhibit anti-inflammatory, antiproliferative, antioxidant properties, and antimicrobial activities. The aim of this study is to evaluate these activities of Lacticaseibacillus paracasei lysates isolated from fermented palm sap. The bacterial cell lysates were prepared via sonication, and their antibacterial activity was assessed against three pathogens using agar well diffusion assay. Antioxidant activity was evaluated using DPPH and ABTS radical scavenging assays. Cytotoxicity and anti-inflammatory activity were determined in LPS-stimulated RAW 264.7 cells by measuring nitric oxide production, secretion, and mRNA level of cytokines (IL-1β, IL-6, and IL-10). Among the lysates, L. paracasei T1901 demonstrated the strongest antibacterial activity against Escherichia coli, Bacillus cereus, and Acinetobacter baumannii. Most lysates exhibited potent antioxidant activity, especially T0601 that showed the highest DPPH and ABTS radical scavenging activities. In LPS-stimulated RAW 264.7 cells, the lysates effectively reduced nitric oxide levels and suppressed the production of pro-inflammatory cytokines (IL-1β and IL-6); simultaneously, the lysates enhanced immunosuppressive cytokine IL-10 secretion. Furthermore, no cytotoxic effect was observed in the lysate-treated RAW 264.7 cells. The study highlights the potential of L. paracasei lysates as multifunctional agents with antibacterial, antioxidant, and anti-inflammatory properties. These findings support their application in developing functional foods or therapeutic agents for managing oxidative stress and inflammatory diseases.
Collapse
Affiliation(s)
- Phoomjai Sornsenee
- Department of Family and Preventive Medicine, Faculty of Medicine, Prince of Songkla University, Songkhla, 90110, Thailand
| | - Nateelak Kooltheat
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Dechawat Wongprot
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80160, Thailand
- College of Graduate Studies, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Pinkanok Suksabay
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80160, Thailand
- College of Graduate Studies, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Tae-Gyu Nam
- Department of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA, Ansan, Gyeonggi-Do, 15588, Republic of Korea
| | - Uttapol Permpoon
- Department of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University ERICA, Ansan, Gyeonggi-Do, 15588, Republic of Korea
| | - Phanvasri Saengsuwan
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Chonticha Romyasamit
- Department of Medical Technology, School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80160, Thailand.
- Research Center in Tropical Pathobiology, Walailak University, Thasala District, Nakhon Si Thammarat, Thailand.
- Center of Excellence in Innovation of Essential Oil and Bioactive Compounds, Walailak University, Nakhon Si Thammarat, 80160, Thailand.
| |
Collapse
|
42
|
Kubbara EA, Bolad A, Malibary H. Advances in Liposomal Interleukin and Liposomal Interleukin Gene Therapy for Cancer: A Comprehensive Review of Preclinical Studies. Pharmaceutics 2025; 17:383. [PMID: 40143046 PMCID: PMC11945541 DOI: 10.3390/pharmaceutics17030383] [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: 12/13/2024] [Revised: 01/16/2025] [Accepted: 01/27/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Preclinical studies on liposomal interleukin (IL) therapy demonstrate considerable promise in cancer treatment. This review explores the achievements, challenges, and future potential of liposomal IL encapsulation, focusing on preclinical studies. METHODS A structured search was conducted using the PubMed and Web of Science databases with the following search terms and Boolean operators: ("liposomal interleukin" OR "liposome-encapsulated interleukin") AND ("gene therapy" OR "gene delivery") AND ("cancer" OR "tumor" OR "oncology") AND ("pre-clinical studies" OR "animal models" OR "in vitro studies". RESULTS Liposomal IL-2 formulations are notable for enhancing delivery and retention at tumor sites. Recombinant human interleukin (rhIL-2) adsorbed onto small liposomes (35-50 nm) substantially reduces metastases in murine models. Hepatic metastasis models demonstrate superior efficacy of liposomal IL-2 over free IL-2 by enhancing immune responses, particularly in the liver. Localized delivery strategies, including nebulized liposomal IL-2 in canine pulmonary metastases and intrathoracic administration in murine sarcoma models, reduce systemic toxicity while promoting immune activation and tumor regression. Liposomal IL gene therapy, delivering cytokine genes directly to tumor sites, represents a notable advancement. Combining IL-2 gene therapy with other cytokines, including IL-6 or double-stranded RNA adjuvants, synergistically enhances macrophage and T-cell activation. Liposomal IL-4, IL-6, and IL-21 therapies show potential across various tumor types. Pairing liposomal IL-2 with chemotherapy or immune agents improves remission and survival. Innovative strategies, including PEGylation and ligand-targeted systems, optimize delivery, release, and therapeutic outcomes. CONCLUSIONS Utilizing immune-stimulatory ILs through advanced liposomal delivery and gene therapy establishes a strong foundation for advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Eman A. Kubbara
- Clinical Biochemistry Department, Faculty of Medicine, Rabigh Branch, King Abdulaziz University, Rabigh 21911, Saudi Arabia
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Al-Neelain University, Khartoum 11121, Sudan
| | - Ahmed Bolad
- Department of Microbiology and Unit of Immunology, Faculty of Medicine, Al-Neelain University, Khartoum 11121, Sudan
| | - Husam Malibary
- Department of Medicine, Faculty of Medicine, King Abdulaziz University, Rabigh 21911, Saudi Arabia
| |
Collapse
|
43
|
Gomaa S, Nassef M, Tabl G, Gabry SE. Immunoenhancing of the anti-cancer therapy and anti-oxidative stress by co-administration of granulocyte-colony stimulating factor-mobilized stem cells or cells derived from bone marrow and/or spleen plus vaccination with chemotherapeutic cyclophosphamide. Immunol Res 2025; 73:62. [PMID: 40091102 DOI: 10.1007/s12026-025-09610-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: 10/24/2024] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
The combination of immunotherapy and chemotherapy, referred to as chemo-immunotherapy, represents a promising regimen for developing new cancer treatments that target the local tumor microenvironment and target tumors in their early stages. However, this approach carries potential risks, including myelo- and immunosuppression, as well as the emergence of chemo-resistant tumor cells. The purpose of this study was to investigate how well mobilizing hematopoietic stem cells (HSCs) work when used alongside chemotherapy and immunotherapy to enhance and modulate the immune response, thereby overcoming immunosuppression and eliminating distant cancer cells. Ehrlich ascetic carcinoma (EAC) tumor-bearing mice were intraperitoneal (i.p.) preconditioned with CTX (4 mg/mouse). EAC-bearing mice that were preconditioned with CTX were intravenous (i.v.) administered with adoptive transferred naive mice-derived bone marrow cells (nBMCs) at 5 × 106 through lateral tail vein (nBMCs group), adoptive transferred tumor-bearing mice-derived bone marrow cells (tBMCs) at 5 × 106 cell/mouse (tBMCs group), a combination of adoptive transferred naïve mice-derived bone marrow cells (nBMCs) and naïve mice-derived splenocytes (nSPs) at 5 × 106 (nBMCs/nSPs group), a combination of adoptive transferred tumor-bearing mice-derived bone marrow cells (tBMCs) and tumor-bearing mice derived-splenocytes (tSPs) at 5 × 106 cell/mouse (tBMCs/tSPs group), or G-CSF administrated subcutaneously (s.c.) at 5 µg/mouse (G-CSF group). Subsequently, all mice groups were vaccinated with tumor lysate at a dosage of 100 µg/mouse. Treating EAC tumor-bearing mice with G-CSF, adoptive transferred nBMCs, adoptive transferred tBMCs, adoptive transferred nBMCs/nSPs, adoptive transferred tBMCs/tSPs, resulted in a significantly enhanced anti-tumor effect that was evidenced by increased anti-proliferative activity and growth inhibition against EAC tumor cells, increased necrosis and apoptosis rates among EAC tumor cells, restricted tumor growth in EAC tumor-bearing mice, and reduced levels of carcinoembryonic antigen (CEA) tumor marker. Furthermore, there was an improvement in serum levels of antioxidant enzyme superoxide dismutase (SOD) and malondialdehyde (MDA) in EAC tumor-bearing mice receiving G-CSF, adoptive transferred tBMCs, adoptive transferred nBMCs/nSPs, and adoptive transferred tBMCs/tSPs. Notably, this treatment regimen ameliorates liver and kidney damage associated with CTX administration in EA tumor-bearing mice. The integration of G-CSF-mobilized HSCs, adoptive transferred nBMCs, adoptive transferred tBMCs, adoptive transferred nBMCs/nSPs combination, and adoptive transferred tBMCs/tSPs combination may yield powerful anti-cancer therapy, thereby facilitating more effective anti-tumor immunotherapy strategies when align with anti-tumor responses. This research may propose a novel therapeutic approach that combines chemotherapy and immunotherapy for addressing early-stage cancer. Further research is necessary to connect the biomedical application and heterogeneity of human tumors and immune systems of this regimen to both diagnostic and therapeutic methodologies.
Collapse
Affiliation(s)
- Soha Gomaa
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt.
| | - Mohamed Nassef
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt
| | - Ghada Tabl
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt
| | - Shaimaa El Gabry
- Department of Zoology, Science Faculty, University of Tanta, 31527, Tanta, Egypt
| |
Collapse
|
44
|
Dai X, Fan Y, Zhao X. Systemic lupus erythematosus: updated insights on the pathogenesis, diagnosis, prevention and therapeutics. Signal Transduct Target Ther 2025; 10:102. [PMID: 40097390 PMCID: PMC11914703 DOI: 10.1038/s41392-025-02168-0] [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: 09/04/2024] [Revised: 11/26/2024] [Accepted: 01/26/2025] [Indexed: 03/19/2025] Open
Abstract
Systemic lupus erythematosus (SLE) is a chronic inflammatory illness with heterogeneous clinical manifestations covering multiple organs. Diversified types of medications have been shown effective for alleviating SLE syndromes, ranging from cytokines, antibodies, hormones, molecular inhibitors or antagonists, to cell transfusion. Drugs developed for treating other diseases may benefit SLE patients, and agents established as SLE therapeutics may be SLE-inductive. Complexities regarding SLE therapeutics render it essential and urgent to identify the mechanisms-of-action and pivotal signaling axis driving SLE pathogenesis, and to establish innovative SLE-targeting approaches with desirable therapeutic outcome and safety. After introducing the research history of SLE and its epidemiology, we categorized primary determinants driving SLE pathogenesis by their mechanisms; combed through current knowledge on SLE diagnosis and grouped them by disease onset, activity and comorbidity; introduced the genetic, epigenetic, hormonal and environmental factors predisposing SLE; and comprehensively categorized preventive strategies and available SLE therapeutics according to their functioning mechanisms. In summary, we proposed three mechanisms with determinant roles on SLE initiation and progression, i.e., attenuating the immune system, restoring the cytokine microenvironment homeostasis, and rescuing the impaired debris clearance machinery; and provided updated insights on current understandings of SLE regarding its pathogenesis, diagnosis, prevention and therapeutics, which may open an innovative avenue in the fields of SLE management.
Collapse
Affiliation(s)
- Xiaofeng Dai
- National Local Joint Engineering Research Center for Precision Surgery & Regenerative Medicine, Shaanxi Provincial Center for Regenerative Medicine and Surgical Engineering, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China.
| | - Yuting Fan
- Tissue Engineering and Stem Cell Experiment Center, Tumor Immunotherapy Technology Engineering Research Center, Department of Immunology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, P. R. China
- Department of Gastroenterology, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550001, P. R. China
| | - Xing Zhao
- Tissue Engineering and Stem Cell Experiment Center, Tumor Immunotherapy Technology Engineering Research Center, Department of Immunology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, P. R. China.
| |
Collapse
|
45
|
El-Demerdash FM, Al Mhanna AB, El-Sayed RA, Mohamed TM, Salem MM. Use of Nigella sativa silver nanocomposite as an alternative therapy against thioacetamide nephrotoxicity. GENES & NUTRITION 2025; 20:6. [PMID: 40087564 PMCID: PMC11909921 DOI: 10.1186/s12263-025-00766-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/19/2025] [Indexed: 03/17/2025]
Abstract
Nigella sativa (N. sativa) L. (Ranunculaceae), commonly referred to as black cumin, has a long history of usage as an herbal remedy. It has been utilized conventionally and in clinical settings to treat various illnesses. Six groups of male Wister rats were randomly selected as Gp I, represented as control; Gp II administered N. sativa aqueous extract (NSAE); 200 mg/kg/d, Gp III received N. sativa silver nanocomposite (NS-Ag-NC); 0.25 mg/kg/d; Gp IV administered thioacetamide (TAA);100 mg/kg; thrice weekly and Gps V and VI administered NSAE and NS-Ag-NC with TAA for six weeks, respectively. Findings showed that GC-MS analysis of NSAE has a high concentration of phytochemicals with strong antioxidant activity. Results revealed that TAA administration elevated TBARS, H2O2, PCC, NO levels, kidney function parameters, LDH activity, and up-regulated TNF-α, IL-1β, NF-kβ, and COX-2 gene expressions. In contrast, enzymatic and non-enzymatic antioxidants and ALP activity were extensively diminished. Also, severe abnormalities in lipid profile, hematological parameters, and histopathological features were noted. On the other hand, the administration of NSAE or NS-Ag-NC followed by TAA intoxication reduces renal impairment, restores the antioxidant system, and downregulates the expression of TNF-α, IL-1β, NF-kβ, and COX-2 genes in rats' renal tissues. Collectively, NS-Ag-NC has more prevalent nephroprotective impacts than NSAE and can adjust the oxidant/antioxidant pathways besides their anti-inflammatory efficacy against TAA toxicity.
Collapse
Affiliation(s)
- Fatma M El-Demerdash
- Department of Environmental Studies, Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21526, Egypt.
| | - Ansam B Al Mhanna
- Department of Environmental Studies, Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21526, Egypt
| | - Raghda A El-Sayed
- Department of Environmental Studies, Institute of Graduate Studies and Research, Alexandria University, Alexandria, 21526, Egypt
| | - Tarek M Mohamed
- Biochemistry Division, Department of Chemistry, Tanta University, Tanta, 31527, Egypt
| | - Maha M Salem
- Biochemistry Division, Department of Chemistry, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
46
|
Trivadila T, Iswantini D, Rahminiwati M, Rafi M, Salsabila AP, Sianipar RNR, Indariani S, Murni A. Herbal Immunostimulants and Their Phytochemicals: Exploring Morinda citrifolia, Echinacea purpurea, and Phyllanthus niruri. PLANTS (BASEL, SWITZERLAND) 2025; 14:897. [PMID: 40265854 PMCID: PMC11945065 DOI: 10.3390/plants14060897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/01/2025] [Accepted: 03/06/2025] [Indexed: 04/24/2025]
Abstract
The rising prevalence of infectious diseases and immune-related disorders underscores the need for effective and accessible therapeutic solutions. Herbal immunostimulants derived from medicinal plants offer promising alternatives, enhancing immune responses with lower toxicity and fewer side effects than synthetic drugs. This review explores the immunostimulatory potential of Morinda citrifolia, Echinacea purpurea, and Phyllanthus niruri, focusing on their bioactive compounds, mechanisms of action, and therapeutic relevance. These plants modulate innate and adaptive immune responses by activating macrophages, dendritic cells, and lymphocytes while regulating cytokine production to maintain immune homeostasis. Their immunomodulatory effects are linked to key signaling pathways, including NF-κB, MAPK, and JAK/STAT. In vitro and in vivo studies highlight their potential to strengthen immune responses and control inflammation, making them promising candidates for managing infectious and immune-related diseases. However, further research is needed to standardize formulations, determine optimal dosages, and validate safety and efficacy in clinical settings. Addressing these gaps will support the integration of herbal immunostimulants into evidence-based healthcare as sustainable and accessible immune-enhancing strategies.
Collapse
Affiliation(s)
- Trivadila Trivadila
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University, Bogor 16680, West Java, Indonesia; (T.T.); (M.R.); (A.P.S.); (R.N.R.S.)
- Tropical Biopharmaca Research Center, IPB University, Bogor 16128, West Java, Indonesia; (M.R.); (S.I.); (A.M.)
| | - Dyah Iswantini
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University, Bogor 16680, West Java, Indonesia; (T.T.); (M.R.); (A.P.S.); (R.N.R.S.)
- Tropical Biopharmaca Research Center, IPB University, Bogor 16128, West Java, Indonesia; (M.R.); (S.I.); (A.M.)
| | - Min Rahminiwati
- Tropical Biopharmaca Research Center, IPB University, Bogor 16128, West Java, Indonesia; (M.R.); (S.I.); (A.M.)
- School of Veterinary Medicine and Biomedical Sciences, IPB University, Bogor 16680, West Java, Indonesia
| | - Mohamad Rafi
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University, Bogor 16680, West Java, Indonesia; (T.T.); (M.R.); (A.P.S.); (R.N.R.S.)
- Tropical Biopharmaca Research Center, IPB University, Bogor 16128, West Java, Indonesia; (M.R.); (S.I.); (A.M.)
| | - Adisa Putri Salsabila
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University, Bogor 16680, West Java, Indonesia; (T.T.); (M.R.); (A.P.S.); (R.N.R.S.)
| | - Rut Novalia Rahmawati Sianipar
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, IPB University, Bogor 16680, West Java, Indonesia; (T.T.); (M.R.); (A.P.S.); (R.N.R.S.)
| | - Susi Indariani
- Tropical Biopharmaca Research Center, IPB University, Bogor 16128, West Java, Indonesia; (M.R.); (S.I.); (A.M.)
| | - Anggia Murni
- Tropical Biopharmaca Research Center, IPB University, Bogor 16128, West Java, Indonesia; (M.R.); (S.I.); (A.M.)
| |
Collapse
|
47
|
Elkotamy MS, Elgohary MK, Alkabbani MA, Binjubair FA, Alanazi MM, Alsulaimany M, Al-Rashood ST, Ghabbour HA, Abdel-Aziz HA. Design, synthesis and biological evaluation of pyrazolo[3,4- b]pyridine derivatives as dual CDK2/PIM1 inhibitors with potent anti-cancer activity and selectivity. J Biomol Struct Dyn 2025:1-25. [PMID: 40079180 DOI: 10.1080/07391102.2025.2475233] [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: 10/20/2024] [Accepted: 01/31/2025] [Indexed: 03/14/2025]
Abstract
The discovery of novel, selective inhibitors targeting CDK2 and PIM1 kinases, which regulate cell survival, proliferation, and treatment resistance, is crucial for advancing cancer therapy. This study reports the design, synthesis, and biological evaluation of three novel pyrazolo[3,4-b]pyridine derivatives (6a-c), confirmed via spectral analyses. These compounds were assessed for anti-cancer activity against breast, colon, liver, and cervical cancers using the MTT assay. Among the tested compounds, 6b exhibited superior efficacy, with higher selectivity indices for HCT-116 (15.05) and HepG2 (9.88) compared to the reference drug staurosporine. Mechanistic studies revealed that 6b induced apoptosis (63.04-fold increase) and arrested the cell cycle at the G0-G1 phase, highlighting its anti-proliferative effects. In an in-vivo solid Ehrlich carcinoma (SEC) mouse model, compound 6b significantly reduced tumor weight and volume, exceeding the efficacy of doxorubicin. Additionally, 6b potently inhibited CDK2 and PIM1 kinases (IC50: 0.27 and 0.67 µM, respectively) and reduced tumor-promoting TNF-alpha expression, as confirmed by histopathological and immunohistochemical studies. Computational analyses, including molecular docking, molecular dynamics simulations, and DFT calculations, provided insights into the binding stability and interaction mechanisms of 6b with CDK2 and PIM1, while in-silico pharmacokinetic and toxicity evaluations confirmed its favorable drug-like profile and safety. This study highlights compound 6b as a promising dual CDK2/PIM1 inhibitor with potent anti-cancer activity and selectivity, paving the way for its further optimization and development as a lead molecule in cancer therapy.
Collapse
Affiliation(s)
- Mahmoud S Elkotamy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Egypt
| | - Mohamed K Elgohary
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Egypt
| | | | - Faizah A Binjubair
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Manal M Alanazi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Marwa Alsulaimany
- Department of Pharmacognosy and Pharmaceutical Chemistry, College of Pharmacy, Taibah University, Medina, Saudi Arabia
| | - Sara T Al-Rashood
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hazem A Ghabbour
- School of Health and Biomedical Sciences, RMIT University, Melbourne, Australia
| | - Hatem A Abdel-Aziz
- Applied Organic Chemistry Department, National Research Center, Dokki, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| |
Collapse
|
48
|
Turlej E, Domaradzka A, Radzka J, Drulis-Fajdasz D, Kulbacka J, Gizak A. Cross-Talk Between Cancer and Its Cellular Environment-A Role in Cancer Progression. Cells 2025; 14:403. [PMID: 40136652 PMCID: PMC11940884 DOI: 10.3390/cells14060403] [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: 01/30/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025] Open
Abstract
The tumor microenvironment is a dynamic and complex three-dimensional network comprising the extracellular matrix and diverse non-cancerous cells, including fibroblasts, adipocytes, endothelial cells and various immune cells (lymphocytes T and B, NK cells, dendritic cells, monocytes/macrophages, myeloid-derived suppressor cells, and innate lymphoid cells). A constantly and rapidly growing number of studies highlight the critical role of these cells in shaping cancer survival, metastatic potential and therapy resistance. This review provides a synthesis of current knowledge on the modulating role of the cellular microenvironment in cancer progression and response to treatment.
Collapse
Affiliation(s)
- Eliza Turlej
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Aleksandra Domaradzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Justyna Radzka
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Dominika Drulis-Fajdasz
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| | - Julita Kulbacka
- Departament of Molecular and Cellular Biology, Faculty of Pharmacy, Wrocław Medical University, Borowska 211A, 50-556 Wrocław, Poland;
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine, LT-08406 Vilnius, Lithuania
| | - Agnieszka Gizak
- Departament of Molecular Physiology and Neurobiology, University of Wrocław, ul. Sienkiewicza 21, 50-335 Wrocław, Poland; (E.T.); (A.D.); (J.R.)
| |
Collapse
|
49
|
Iosub G, Lungescu IA, Bîrcă AC, Niculescu AG, Balaure PC, Constantinescu S, Mihaiescu B, Rădulescu DM, Grumezescu AM, Hudiță A, Neacșu IA, Rădulescu AR. New Three Dimensional-Printed Polyethylene Terephthalate Glycol Liners for Hip Joint Endoprostheses: A Bioactive Platform for Bone Regeneration. MATERIALS (BASEL, SWITZERLAND) 2025; 18:1206. [PMID: 40141489 PMCID: PMC11944038 DOI: 10.3390/ma18061206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 02/18/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025]
Abstract
Osteoporosis and bone defects are commonly observed in postmenopausal women, often linked to decreased folic acid levels, which play a crucial role in bone metabolism and regeneration. This study investigates 3D-printed polyethylene terephthalate glycol (PETG)-based porous scaffolds impregnated with chitosan (CS), hydroxyapatite (HAp), and folic acid (FA) for bone tissue engineering applications. The PETG-CS scaffold serves as the primary structural framework, with HAp incorporated to enhance bioactivity through its osteoconductive and osteoinductive properties. FA was included to address potential deficiencies in bone quality and to stimulate cellular differentiation. The scaffolds were fabricated using precise 3D printing techniques, yielding structures with controlled porosity. Physicochemical analyses confirmed the successful integration of HAp and FA into the PETG-CS matrix. Biological evaluations using preosteoblast cell lines demonstrated enhanced cell viability, proliferation, and biocompatibility of the scaffolds. These findings highlight the promising applications of PETG-CS-HAp-FA scaffolds in bone tissue engineering, providing a platform for future investigations into personalized regenerative therapies.
Collapse
Affiliation(s)
- Gheorghe Iosub
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania; (G.I.); (S.C.); (A.R.R.)
| | - Ioana-Alexandra Lungescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania; (I.-A.L.); (A.C.B.); (A.-G.N.); (A.M.G.); (I.A.N.)
| | - Alexandra Cătălina Bîrcă
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania; (I.-A.L.); (A.C.B.); (A.-G.N.); (A.M.G.); (I.A.N.)
| | - Adelina-Gabriela Niculescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania; (I.-A.L.); (A.C.B.); (A.-G.N.); (A.M.G.); (I.A.N.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 90-92 Panduri, 050663 Bucharest, Romania; (B.M.); (A.H.)
| | - Paul Catalin Balaure
- Organic Chemistry Department, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania
| | - Sorin Constantinescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania; (G.I.); (S.C.); (A.R.R.)
| | - Bogdan Mihaiescu
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 90-92 Panduri, 050663 Bucharest, Romania; (B.M.); (A.H.)
| | - Dragoș Mihai Rădulescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania; (G.I.); (S.C.); (A.R.R.)
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania; (I.-A.L.); (A.C.B.); (A.-G.N.); (A.M.G.); (I.A.N.)
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 90-92 Panduri, 050663 Bucharest, Romania; (B.M.); (A.H.)
| | - Ariana Hudiță
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 90-92 Panduri, 050663 Bucharest, Romania; (B.M.); (A.H.)
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania
| | - Ionela Andreea Neacșu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania; (I.-A.L.); (A.C.B.); (A.-G.N.); (A.M.G.); (I.A.N.)
| | - Adrian Radu Rădulescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania; (G.I.); (S.C.); (A.R.R.)
| |
Collapse
|
50
|
Zhang W, Wang R, Guo R, Yi Z, Wang Y, Wang H, Li Y, Li X, Song J. The multiple biological activities of hyperoside: from molecular mechanisms to therapeutic perspectives in neoplastic and non-neoplastic diseases. Front Pharmacol 2025; 16:1538601. [PMID: 40098612 PMCID: PMC11911483 DOI: 10.3389/fphar.2025.1538601] [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: 12/03/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
In recent years, hyperoside (quercetin 3-O-β-D-galactopyranoside) has garnered significant attention due to its diverse biological effects, which include vasoprotective, antioxidant, anti-inflammatory, and anti-tumor properties. Notably, hyperoside has shown remarkable potential in cancer therapy by targeting multiple mechanisms; it induces apoptosis, inhibits proliferation, blocks angiogenesis, and reduces the metastatic potential of cancer cells. Furthermore, hyperoside enhances the sensitivity of cancer cells to chemotherapy by modulating key signaling pathways. Beyond neoplastic diseases, hyperoside also presents promising therapeutic applications in managing non-cancerous conditions such as diabetes, Alzheimer's disease, and pulmonary fibrosis. This review comprehensively examines the molecular mechanisms underlying hyperoside's anti-cancer effects and highlights its role in the treatment of cancers, including lung and colorectal cancers. Additionally, it explores the latest research on hyperoside's potential in addressing non-neoplastic conditions, such as pulmonary fibrosis, diabetes, and Parkinson's disease. By summarizing current findings, this review underscores the unique therapeutic value of hyperoside and its potential as a multifunctional treatment in both neoplastic and non-neoplastic contexts.
Collapse
Affiliation(s)
- Weisong Zhang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Rui Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Rongqi Guo
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Zhongquan Yi
- Central Laboratory, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Yihao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Hao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Yangyang Li
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
- Medical School of Nantong University, Nantong, China
| | - Xia Li
- Department of General Medicine, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| | - Jianxiang Song
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, China
| |
Collapse
|