1
|
Liu S, Ma Z. The role of cannabinoid-mediated signaling pathways and mechanisms in brain disorders. Cell Signal 2025; 128:111653. [PMID: 39952540 DOI: 10.1016/j.cellsig.2025.111653] [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: 12/12/2024] [Revised: 01/17/2025] [Accepted: 02/07/2025] [Indexed: 02/17/2025]
Abstract
Cannabinoids play significant roles in the central nervous system (CNS), but cannabinoid-mediated physiopathological functions are not elaborated. Cannabinoid receptors (CBRs) mediate functions that include the regulation of neuroinflammation, oxidative stress, apoptosis, autophagy, and neurogenesis. Microglia are the primary immune cells responsible for mediating neuroinflammation in the CNS. Therefore, this article primarily focuses on microglia to summarize the inflammatory pathways mediated by cannabinoids in the CNS, including nuclear factor-κB (NF-κB), NOD-like receptor protein 3 (NLRP3) inflammasome, mitogen-activated protein kinase (MAPK), protein kinase B (Akt), and cAMP-dependent protein kinase (PKA) signaling pathways. Additionally, we provide a table summarizing the role of cannabinoids in various brain diseases. Medical use of cannabinoids has protective effects in preventing and treating brain diseases; however, excessive and repeated use can be detrimental to the CNS. We propose that cannabinoids hold significant potential for preventing and treating brain diseases, including ferroptosis, lactate metabolism, and mitophagy, providing new insights for further research on cannabinoids.
Collapse
Affiliation(s)
- Shunfeng Liu
- School of Basic Medicine, Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disorders, Qingdao University, Qingdao 266071, China
| | - Zegang Ma
- School of Basic Medicine, Qingdao University, Qingdao 266071, China; Institute of Brain Science and Disorders, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
2
|
Melo ESA, Asevedo EA, Duarte-Almeida JM, Nurkolis F, Syahputra RA, Park MN, Kim B, do Couto RO, Ribeiro RIMDA. Mechanisms of Cell Death Induced by Cannabidiol Against Tumor Cells: A Review of Preclinical Studies. PLANTS (BASEL, SWITZERLAND) 2025; 14:585. [PMID: 40006844 PMCID: PMC11859785 DOI: 10.3390/plants14040585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025]
Abstract
Commonly known as marijuana or hemp, Cannabis sativa L. (Cannabaceae), contains numerous active compounds, particularly cannabinoids, which have been extensively studied for their biological activities. Among these, cannabidiol (CBD) stands out for its therapeutic potential, especially given its non-psychotropic effects. This review evaluates the antitumor properties of CBD, highlighting its various mechanisms of action, including the induction of apoptosis, autophagy, and necrosis. By synthesizing findings from in vitro studies on the cell death mechanisms and signaling pathways activated by CBD in various human tumor cell lines, this literature review emphasizes the therapeutic promise of this natural antineoplastic agent. We conducted a comprehensive search of articles in PubMed, Scopus, Springer, Medline, Lilacs, and Scielo databases from 1984 to February 2022. Of the forty-three articles included, the majority (68.18%) reported that CBD activates apoptosis, while 18.18% observed simultaneous apoptosis and autophagy, 9.09% focused on autophagy alone, and 4.54% indicated necrosis. The antitumor effects of CBD appear to be mediated by transient receptor potential cation channels (TRPVs) in endometrial cancer, glioma, bladder cancer, and myeloma, with TRPV1, TRPV2, and TRPV4 playing key roles in activating apoptosis. This knowledge paves the way for innovative therapeutic strategies that may enhance cancer treatment outcomes while minimizing the toxicity and side effects associated with conventional therapies.
Collapse
Affiliation(s)
- Edilene S. A. Melo
- Experimental Pathology Laboratory, Dona Lindu Central-West Campus (CCO), Federal University of São João del-Rei (UFSJ), Sebastião Gonçalves Coelho 400, Chanadour, Divinopolis 35501-296, MG, Brazil; (E.S.A.M.); (E.A.A.)
| | - Estefani A. Asevedo
- Experimental Pathology Laboratory, Dona Lindu Central-West Campus (CCO), Federal University of São João del-Rei (UFSJ), Sebastião Gonçalves Coelho 400, Chanadour, Divinopolis 35501-296, MG, Brazil; (E.S.A.M.); (E.A.A.)
| | - Joaquim Maurício Duarte-Almeida
- Plant Cell Culture Laboratory, Dona Lindu Central-West Campus (CCO), Federal University of São João del-Rei, Sebastião Gonçalves Coelho 400, Chanadour, Divinopolis 35501-296, MG, Brazil;
| | - Fahrul Nurkolis
- Department of Biological Sciences, Faculty of Sciences and Technology, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta 55281, Indonesia;
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan 20155, Indonesia;
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.N.P.); (B.K.)
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; (M.N.P.); (B.K.)
| | - Renê Oliveira do Couto
- Pharmaceutical Development Laboratory, Dona Lindu Central-West Campus (CCO), Federal University of São João del-Rei, Sebastião Gonçalves Coelho 400, Chanadour, Divinopolis 35501-296, MG, Brazil;
| | - Rosy Iara Maciel de A. Ribeiro
- Experimental Pathology Laboratory, Dona Lindu Central-West Campus (CCO), Federal University of São João del-Rei (UFSJ), Sebastião Gonçalves Coelho 400, Chanadour, Divinopolis 35501-296, MG, Brazil; (E.S.A.M.); (E.A.A.)
| |
Collapse
|
3
|
Mesas C, Moreno J, Doello K, Peña M, López-Romero JM, Prados J, Melguizo C. Cannabidiol effects in stem cells: A systematic review. Biofactors 2025; 51:e2148. [PMID: 39653426 DOI: 10.1002/biof.2148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/22/2024] [Indexed: 12/29/2024]
Abstract
Stem cells play a critical role in human tissue regeneration and repair. In addition, cancer stem cells (CSCs), subpopulations of cancer cells sharing similar characteristics as normal stem cells, are responsible for tumor metastasis and resistance to chemo- and radiotherapy and to tumor relapse. Interestingly, all stem cells have cannabinoid receptors, such as cannabidiol (CBD), that perform biological functions. The aim of this systematic review was to analyze the effect of CBD on both somatic stem cells (SSCs) and CSCs. Of the 276 articles analyzed, 38 were selected according to the inclusion and exclusion criteria. A total of 27 studied the effect of CBD on SSCs, finding that 44% focused on CBD differentiation effect and 56% on its protective activity. On the other hand, 11 articles looked at the effect of CBD on CSCs, including glioblastoma (64%), lung cancer (27%), and breast cancer (only one article). Our results showed that CBD exerted a differentiating and protective effect on SCCs. In addition, this molecule demonstrated an antiproliferative effect on some CSCs, although most of the analyses were performed in vitro. Therefore, although in vivo studies should be necessary to justify its clinical use, CBD and its receptors could be a specific target to act on both SSCs and CSCs.
Collapse
Affiliation(s)
- Cristina Mesas
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (ibs.GRANADA), Granada, Spain
| | - Javier Moreno
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Kevin Doello
- Instituto de Investigación Biosanitaria de Granada, (ibs.GRANADA), Granada, Spain
- Service of Medical Oncology, Hospital Virgen de las Nieves, Granada, Spain
| | - Mercedes Peña
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (ibs.GRANADA), Granada, Spain
| | - Juan M López-Romero
- Department of Organic Chemistry, Faculty of Sciences, University of Malaga, Málaga, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria de Granada, (ibs.GRANADA), Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| |
Collapse
|
4
|
Billi M, Pagano S, Pancrazi GL, Valenti C, Bruscoli S, Di Michele A, Febo M, Grignani F, Marinucci L. DNA damage and cell death in human oral squamous cell carcinoma cells: The potential biological effects of cannabidiol. Arch Oral Biol 2025; 169:106110. [PMID: 39426313 DOI: 10.1016/j.archoralbio.2024.106110] [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: 07/08/2024] [Revised: 10/06/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
OBJECTIVE The present study examined the in vitro effects on oral squamous cell carcinoma cells (HSC-3) of cannabidiol (CBD), the main chemical component of Cannabis, proposed as a novel adjuvant therapy in the treatment of cancers. DESIGN Cell viability (MTT assay), morphology (SEM), apoptosis and cell cycle (flow cytometry), and DNA damage (phospho-γ-H2AX immunofluorescence) were evaluated. Cytotoxicity was evaluated with concentrations between 100 µM and 1 µM, and two concentrations were selected for subsequent analysis: 25 µM, as toxic dose, and 6.25 µM, as non-toxic. RESULTS CBD caused a dose- and time-dependent reduction in viability of 64 %, 96 %, and 99 % with 25 µM, 50 µM and 100 µM, respectively, after 72 h (p < 0.001), cell cycle arrest in G0-G1 phase with increased apoptosis in particular at 72 h for 25 µM (p < 0.001), significant morphological alterations with 25 µM, still present even at 6.25 µM, and significantly increased cell damage considering a significant increase in the percentage of highly positive cells (5 phosphorylated γH2AX foci), which is around 29 % for 25 µM and 19 % for 6.25 µM after 24 h. CONCLUSIONS CBD inhibits oral cancer growth causing DNA damage. In general, induced cell cytotoxicity appears to be dose- and time-related. Doses of CBD ≥25 μM showed a high reduction in viability. CBD could possibly represent a new therapeutic molecule for its cytotoxic effects against oral squamous cell carcinoma. The mechanism involved in the suppressive effect caused by CBD needs further investigation.
Collapse
Affiliation(s)
- Monia Billi
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, S. Andrea delle Fratte, Perugia 06156, Italy.
| | - Stefano Pagano
- Department of Medicine and Surgery, Faculty of Dentistry, University of Perugia, S. Andrea delle Fratte, Perugia 06156, Italy.
| | - Gian Luca Pancrazi
- Department of Medicine and Surgery, Faculty of Dentistry, University of Perugia, S. Andrea delle Fratte, Perugia 06156, Italy.
| | - Chiara Valenti
- Department of Medicine and Surgery, Faculty of Dentistry, University of Perugia, S. Andrea delle Fratte, Perugia 06156, Italy; CISAS "Giuseppe Colombo", University of Padua, Via Venezia, 15, Padua 35131, Italy.
| | - Stefano Bruscoli
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, S. Andrea delle Fratte, Perugia 06156, Italy.
| | - Alessandro Di Michele
- Department of Physics and Geology, University of Perugia, Via Pascoli, Perugia 06123, Italy.
| | - Marta Febo
- Department of Medicine and Surgery, Section of Pharmacology, University of Perugia, S. Andrea delle Fratte, Perugia 06156, Italy.
| | - Francesco Grignani
- Department of Medicine and Surgery, Section of General Pathology, University of Perugia, S. Andrea delle Fratte, Perugia 06156, Italy.
| | - Lorella Marinucci
- Department of Medicine and Surgery, Section of Biosciences and Medical Embryology, University of Perugia, S. Andrea delle Fratte, Perugia 06156, Italy.
| |
Collapse
|
5
|
Huang C, Zhang L, Shen P, Wu Z, Li G, Huang Y, Ao T, Luo L, Hu C, Wang N, Quzhuo R, Tian L, Huangfu C, Liao Z, Gao Y. Cannabidiol mitigates radiation-induced intestine ferroptosis via facilitating the heterodimerization of RUNX3 with CBFβ thereby promoting transactivation of GPX4. Free Radic Biol Med 2024; 222:288-303. [PMID: 38830513 DOI: 10.1016/j.freeradbiomed.2024.05.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/28/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
Radiation enteritis remains a major challenge for radiotherapy against abdominal and pelvic malignancies. Nevertheless, there is no approved effective therapy to alleviate irradiation (IR)-induced gastrointestinal (GI) toxicity. In the current study, Cannabidiol (CBD) was found to mitigate intestinal injury by GPX4-mediated ferroptosis resistance upon IR exposure. RNA-sequencing was employed to investigate the underlying mechanism involved in the radio-protective effect of CBD, wherein runt-related transcription factor 3 (RUNX3) and its target genes were changed significantly. Further experiment showed that the transactivation of GPX4 triggered by the direct binding of RUNX3 to its promoter region, or by stimulating the transcriptional activity of NF-κB via RUNX3-mediated LILRB3 upregulation was critical for the anti-ferroptotic effect of CBD upon IR injury. Specially, CBD was demonstrated to be a molecular glue skeleton facilitating the heterodimerization of RUNX3 with its transcriptional chaperone core-biding factor β (CBFβ) thereby promoting their nuclear localization and the subsequent transactivation of GPX4 and LILRB3. In short, our study provides an alternative strategy to counteract IR-induced enteritis during the radiotherapy on abdominal/pelvic neoplasms.
Collapse
Affiliation(s)
- Congshu Huang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Liangliang Zhang
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Pan Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Zekun Wu
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Gaofu Li
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yijian Huang
- Senior Department of Orthopedics, The Fourth Medical Center of PLA General Hospital, Beijing, 100048, China
| | - Ting Ao
- College of Pharmaceutical Science, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Lin Luo
- School of Nursing, Capital Medical University, Beijing, 100069, China
| | - Changkun Hu
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ningning Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Renzeng Quzhuo
- Department of General Internal Medicine, Naqu People's Hospital, Nagqu, Xizang Autonomous Region, 852007, China
| | - Lishan Tian
- Navy Qingdao Special Service Recuperation Center, Qingdao, 266071, China
| | - Chaoji Huangfu
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Zebin Liao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Yue Gao
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| |
Collapse
|
6
|
Cheng J, Li M, Motta E, Barci D, Song W, Zhou D, Li G, Zhu S, Yang A, Vaillant BD, Imhof A, Forné I, Spiegl-Kreinecker S, Zhang N, Katayama H, Bhat KPL, Flüh C, Kälin RE, Glass R. Myeloid cells coordinately induce glioma cell-intrinsic and cell-extrinsic pathways for chemoresistance via GP130 signaling. Cell Rep Med 2024; 5:101658. [PMID: 39053460 PMCID: PMC11384956 DOI: 10.1016/j.xcrm.2024.101658] [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: 11/28/2023] [Revised: 05/10/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024]
Abstract
The DNA damage response (DDR) and the blood-tumor barrier (BTB) restrict chemotherapeutic success for primary brain tumors like glioblastomas (GBMs). Coherently, GBMs almost invariably relapse with fatal outcomes. Here, we show that the interaction of GBM and myeloid cells simultaneously induces chemoresistance on the genetic and vascular levels by activating GP130 receptor signaling, which can be addressed therapeutically. We provide data from transcriptomic and immunohistochemical screens with human brain material and pharmacological experiments with a humanized organotypic GBM model, proteomics, transcriptomics, and cell-based assays and report that nanomolar concentrations of the signaling peptide humanin promote temozolomide (TMZ) resistance through DDR activation. GBM mouse models recapitulating intratumoral humanin release show accelerated BTB formation. GP130 blockade attenuates both DDR activity and BTB formation, resulting in improved preclinical chemotherapeutic efficacy. Altogether, we describe an overarching mechanism for TMZ resistance and outline a translatable strategy with predictive markers to improve chemotherapy for GBMs.
Collapse
Affiliation(s)
- Jiying Cheng
- Neurosurgical Research, University Hospital, LMU Munich, Munich, Germany; Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Min Li
- Neurosurgical Research, University Hospital, LMU Munich, Munich, Germany
| | - Edyta Motta
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin-Buch, Germany
| | - Deivi Barci
- Neurosurgical Research, University Hospital, LMU Munich, Munich, Germany
| | - Wangyang Song
- Neurosurgical Research, University Hospital, LMU Munich, Munich, Germany
| | - Ding Zhou
- Neurosurgical Research, University Hospital, LMU Munich, Munich, Germany
| | - Gen Li
- Neurosurgical Research, University Hospital, LMU Munich, Munich, Germany
| | - Sihan Zhu
- Neurosurgical Research, University Hospital, LMU Munich, Munich, Germany
| | - Anru Yang
- Neurosurgical Research, University Hospital, LMU Munich, Munich, Germany
| | - Brian D Vaillant
- Department of Neurology, Dell Medical School, The University of Texas at Austin, Austin, TX, USA
| | - Axel Imhof
- Protein Analysis Unit, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, Martinsried, Germany
| | - Ignasi Forné
- Protein Analysis Unit, Biomedical Center (BMC), Faculty of Medicine, Ludwig-Maximilians-University (LMU) Munich, Martinsried, Germany
| | - Sabine Spiegl-Kreinecker
- Department of Neurosurgery, Medical Faculty, Johannes Kepler University Linz, Linz, Austria; Clinical Research Institute for Neurosciences, Johannes Kepler University Linz, Linz, Austria
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, P.R. China
| | - Hiroshi Katayama
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Charlotte Flüh
- Department of Neurosurgery, University Medical Center Göttingen, Göttingen, Germany
| | - Roland E Kälin
- Neurosurgical Research, University Hospital, LMU Munich, Munich, Germany; Department of Neurosurgery, Medical Faculty, Johannes Kepler University Linz, Linz, Austria
| | - Rainer Glass
- Neurosurgical Research, University Hospital, LMU Munich, Munich, Germany; German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and University Hospital Munich, Munich, Germany; Institute of Surgical Research at the Walter Brendel Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany.
| |
Collapse
|
7
|
Liu F, Wu Q, Liu Q, Chen B, Liu X, Pathak JL, Watanabe N, Li J. Dental pulp stem cells-derived cannabidiol-treated organoid-like microspheroids show robust osteogenic potential via upregulation of WNT6. Commun Biol 2024; 7:972. [PMID: 39122786 PMCID: PMC11315977 DOI: 10.1038/s42003-024-06655-y] [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: 02/13/2023] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Dental pulp stem cells (DPSC) have shown osteogenic and bone regenerative potential. Improving the in situ bone regeneration potential of DPSC is crucial for their application as seed cells during bone defect reconstruction in clinics. This study aimed to develop DPSC-derived organoid-like microspheroids as effective seeds for bone tissue engineering applications. DPSC osteogenic microspheroids (70 μm diameter) were cultured in a polydimethylsiloxane-mold-based agarose-gel microwell-culture-system with or without cannabidiol (CBD)-treatment. Results of in vitro studies showed higher osteogenic differentiation potential of microspheroids compared with 2D-cultured-DPSC. CBD treatment further improved the osteogenic differentiation potential of microspheroids. The effect of CBD treatment in the osteogenic differentiation of microspheroids was more pronounced compared with that of CBD-treated 2D-cultured-DPSC. Microspheroids showed a higher degree of bone regeneration in nude mice calvarial bone defect compared to 2D-cultured-DPSC. CBD-treated microspheroids showed the most robust in situ bone regenerative potential compared with microspheroids or CBD-treated 2D-cultured-DPSC. According to mRNA sequencing, bioinformatic analysis, and confirmation study, the higher osteogenic potential of CBD-treated microspheroids was mainly attributed to WNT6 upregulation. Taken together, DPSC microspheroids have robust osteogenic potential and can effectively translate the effect of in vitro osteoinductive stimulation during in situ bone regeneration, indicating their application potential during bone defect reconstruction in clinics.
Collapse
Affiliation(s)
- Fangqi Liu
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China
| | - Qingqing Wu
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China
| | - Qianwen Liu
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China
| | - Bo Chen
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China
| | - Xintong Liu
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science, Wako, Saitama, 351-0198, Japan
- Bio-Active Compounds Discovery Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama, 351-0198, Japan
| | - Janak L Pathak
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China.
| | - Nobumoto Watanabe
- Chemical Biology Research Group, RIKEN Center for Sustainable Resource Science, Wako, Saitama, 351-0198, Japan
- Bio-Active Compounds Discovery Unit, RIKEN Center for Sustainable Resource Science, Wako, Saitama, 351-0198, Japan
| | - Jiang Li
- School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou, 510182, China.
| |
Collapse
|
8
|
Feng S, Pan Y, Lu P, Li N, Zhu W, Hao Z. From bench to bedside: the application of cannabidiol in glioma. J Transl Med 2024; 22:648. [PMID: 38987805 PMCID: PMC11238413 DOI: 10.1186/s12967-024-05477-0] [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: 05/28/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024] Open
Abstract
Glioma is the most common malignant tumor in central nervous system, with significant health burdens to patients. Due to the intrinsic characteristics of glioma and the lack of breakthroughs in treatment modalities, the prognosis for most patients remains poor. This results in a heavy psychological and financial load worldwide. In recent years, cannabidiol (CBD) has garnered widespread attention and research due to its anti-tumoral, anti-inflammatory, and neuroprotective properties. This review comprehensively summarizes the preclinical and clinical research on the use of CBD in glioma therapy, as well as the current status of nanomedicine formulations of CBD, and discusses the potential and challenges of CBD in glioma therapy in the future.
Collapse
Affiliation(s)
- Shiying Feng
- Department of Oncology, Baotou City Central Hospital, Baotou, 014040, China
- Central Clinical Medical School, Baotou Medical College, Baotou, 014040, China
| | - Yuanming Pan
- Cancer Research Center, Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
| | - Pu Lu
- Department of Oncology, Baotou City Central Hospital, Baotou, 014040, China
| | - Na Li
- Department of Gynecology, Baotou City Central Hospital, Baotou, 014040, China.
| | - Wei Zhu
- Department of Oncology, Baotou City Central Hospital, Baotou, 014040, China.
| | - Zhiqiang Hao
- Department of Oncology, Baotou City Central Hospital, Baotou, 014040, China
| |
Collapse
|
9
|
Lin K, Zhang Y, Shen Y, Xu Y, Huang M, Liu X. Hydrogen Sulfide can Scavenge Free Radicals to Improve Spinal Cord Injury by Inhibiting the p38MAPK/mTOR/NF-κB Signaling Pathway. Neuromolecular Med 2024; 26:26. [PMID: 38907170 DOI: 10.1007/s12017-024-08794-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024]
Abstract
Spinal cord injury (SCI) causes irreversible cell loss and neurological dysfunctions. Presently, there is no an effective clinical treatment for SCI. It can be the only intervention measure by relieving the symptoms of patients such as pain and fever. Free radical-induced damage is one of the validated mechanisms in the complex secondary injury following primary SCI. Hydrogen sulfide (H2S) as an antioxidant can effectively scavenge free radicals, protect neurons, and improve SCI by inhibiting the p38MAPK/mTOR/NF-κB signaling pathway. In this report, we analyze the pathological mechanism of SCI, the role of free radical-mediated the p38MAPK/mTOR/NF-κB signaling pathway in SCI, and the role of H2S in scavenging free radicals and improving SCI.
Collapse
Affiliation(s)
- Kexin Lin
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Yong Zhang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Yanyang Shen
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Yiqin Xu
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Min Huang
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Xuehong Liu
- Department of Histology and Embryology, School of Medicine, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China.
| |
Collapse
|
10
|
Zhang X, Ding T, Yang F, Xu H, Zhang J, Bai Y, Shi Y, Yang J, Chen C, Zhang H. Induced dual-target rebalance simultaneously enhances efficient therapeutical efficacy in tumors. Cell Death Discov 2024; 10:249. [PMID: 38782895 PMCID: PMC11116470 DOI: 10.1038/s41420-024-02018-y] [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: 02/16/2024] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 05/25/2024] Open
Abstract
Multiple gene abnormalities are major drivers of tumorigenesis. NF-κB p65 overactivation and cGAS silencing are important triggers and genetic defects that accelerate tumorigenesis. However, the simultaneous correction of NF-κB p65 and cGAS abnormalities remains to be further explored. Here, we propose a novel Induced Dual-Target Rebalance (IDTR) strategy for simultaneously correcting defects in cGAS and NF-κB p65. By using our IDTR approach, we showed for the first time that oncolytic adenovirus H101 could reactivate silenced cGAS, while silencing GAU1 long noncoding RNA (lncRNA) inhibited NF-κB p65 overactivation, resulting in efficient in vitro and in vivo antitumor efficacy in colorectal tumors. Intriguingly, we further demonstrated that oncolytic adenoviruses reactivated cGAS by promoting H3K4 trimethylation of the cGAS promoter. In addition, silencing GAU1 using antisense oligonucleotides significantly reduced H3K27 acetylation at the NF-κB p65 promoter and inhibited NF-κB p65 transcription. Our study revealed an aberrant therapeutic mechanism underlying two tumor defects, cGAS and NF-κB p65, and provided an alternative IDTR approach based on oncolytic adenovirus and antisense oligonucleotides for efficient therapeutic efficacy in tumors.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- State Key Laboratory of Cardiology and Medical Innovation Center, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, School of Life Science and Technology, Tongji University, Shanghai, 200092, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, 343009, China
- School of Life Science, Jinggangshan University, Ji'an, 343009, China
| | - Tianyi Ding
- State Key Laboratory of Cardiology and Medical Innovation Center, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, School of Life Science and Technology, Tongji University, Shanghai, 200092, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, 343009, China
- School of Life Science, Jinggangshan University, Ji'an, 343009, China
| | - Fan Yang
- State Key Laboratory of Cardiology and Medical Innovation Center, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, School of Life Science and Technology, Tongji University, Shanghai, 200092, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, 343009, China
- School of Life Science, Jinggangshan University, Ji'an, 343009, China
| | - Haowen Xu
- State Key Laboratory of Cardiology and Medical Innovation Center, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, School of Life Science and Technology, Tongji University, Shanghai, 200092, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, 343009, China
- School of Life Science, Jinggangshan University, Ji'an, 343009, China
| | - Jixing Zhang
- State Key Laboratory of Cardiology and Medical Innovation Center, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, School of Life Science and Technology, Tongji University, Shanghai, 200092, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, 343009, China
- School of Life Science, Jinggangshan University, Ji'an, 343009, China
| | - Yiran Bai
- State Key Laboratory of Cardiology and Medical Innovation Center, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, School of Life Science and Technology, Tongji University, Shanghai, 200092, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, 343009, China
- School of Life Science, Jinggangshan University, Ji'an, 343009, China
| | - Yibing Shi
- State Key Laboratory of Cardiology and Medical Innovation Center, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, School of Life Science and Technology, Tongji University, Shanghai, 200092, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, 343009, China
- School of Life Science, Jinggangshan University, Ji'an, 343009, China
| | - Jiaqi Yang
- State Key Laboratory of Cardiology and Medical Innovation Center, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, School of Life Science and Technology, Tongji University, Shanghai, 200092, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, 343009, China
- School of Life Science, Jinggangshan University, Ji'an, 343009, China
| | - Chaoqun Chen
- State Key Laboratory of Cardiology and Medical Innovation Center, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, School of Life Science and Technology, Tongji University, Shanghai, 200092, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, 343009, China
- School of Life Science, Jinggangshan University, Ji'an, 343009, China
| | - He Zhang
- State Key Laboratory of Cardiology and Medical Innovation Center, Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Research Center for Stem Cells, School of Life Science and Technology, Tongji University, Shanghai, 200092, China.
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji'an, 343009, China.
- School of Life Science, Jinggangshan University, Ji'an, 343009, China.
| |
Collapse
|
11
|
Majchrzak-Celińska A, Studzińska-Sroka E. New Avenues and Major Achievements in Phytocompounds Research for Glioblastoma Therapy. Molecules 2024; 29:1682. [PMID: 38611962 PMCID: PMC11013944 DOI: 10.3390/molecules29071682] [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: 02/23/2024] [Revised: 03/22/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024] Open
Abstract
Phytocompounds have been evaluated for their anti-glioblastoma actions for decades, with promising results from preclinical studies but only limited translation into clinics. Indeed, by targeting multiple signaling pathways deregulated in cancer, they often show high efficacy in the in vitro studies, but their poor bioavailability, low tumor accumulation, and rapid clearance compromise their efficacy in vivo. Here, we present the new avenues in phytocompound research for the improvement of glioblastoma therapy, including the ways to enhance the response to temozolomide using phytochemicals, the current focus on phytocompound-based immunotherapy, or the use of phytocompounds as photosensitizers in photodynamic therapy. Moreover, we present new, intensively evaluated approaches, such as chemical modifications of phytochemicals or encapsulation into numerous types of nanoformulations, to improve their bioavailability and delivery to the brain. Finally, we present the clinical trials evaluating the role of phytocompounds or phytocompound-derived drugs in glioblastoma therapy and the less studied phytocompounds or plant extracts that have only recently been found to possess promising anti-glioblastoma properties. Overall, recent advancements in phytocompound research are encouraging; however, only with more 3D glioblastoma models, in vivo studies, and clinical trials it is possible to upgrade the role of phytocompounds in glioblastoma treatment to a satisfactory level.
Collapse
Affiliation(s)
- Aleksandra Majchrzak-Celińska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznan, Poland
| | - Elżbieta Studzińska-Sroka
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznan, Poland;
| |
Collapse
|
12
|
Buchalska B, Kamińska K, Owe-Larsson M, Cudnoch-Jędrzejewska A. Cannabinoids in the treatment of glioblastoma. Pharmacol Rep 2024; 76:223-234. [PMID: 38457018 DOI: 10.1007/s43440-024-00580-x] [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: 12/28/2023] [Revised: 02/16/2024] [Accepted: 02/22/2024] [Indexed: 03/09/2024]
Abstract
Glioblastoma (GBM) is the most prevalent primary malignant tumor of the nervous system. While the treatment of other neoplasms is increasingly more efficacious the median survival rate of GBM patients remains low and equals about 14 months. Due to this fact, there are intensive efforts to find drugs that would help combat GBM. Nowadays cannabinoids are becoming more and more important in the field of cancer and not only because of their properties of antiemetic drugs during chemotherapy. These compounds may have a direct cytotoxic effect on cancer cells. Studies indicate GBM has disturbances in the endocannabinoid system-changes in cannabinoid metabolism as well as in the cannabinoid receptor expression. The GBM cells show expression of cannabinoid receptors 1 and 2 (CB1R and CB2R), which mediate various actions of cannabinoids. Through these receptors, cannabinoids inhibit the proliferation and invasion of GBM cells, along with changing their morphology. Cannabinoids also induce an intrinsic pathway of apoptosis in the tumor. Hence the use of cannabinoids in the treatment of GBM may be beneficial to the patients. So far, studies focusing on using cannabinoids in GBM therapy are mainly preclinical and involve cell lines and mice. The results are promising and show cannabinoids inhibit GBM growth. Several clinical studies are also being carried out. The preliminary results show good tolerance of cannabinoids and prolonged survival after administration of these drugs. In this review, we describe the impact of cannabinoids on GBM and glioma cells in vitro and in animal studies. We also provide overview of clinical trials on using cannabinoids in the treatment of GBM.
Collapse
Affiliation(s)
- Barbara Buchalska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, Warsaw, 02097, Poland
| | - Katarzyna Kamińska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, Warsaw, 02097, Poland.
| | - Maja Owe-Larsson
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, Warsaw, 02097, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Chair and Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Banacha 1b, Warsaw, 02097, Poland
| |
Collapse
|
13
|
Guler A, Hamurcu Z, Ulutabanca H, Cınar V, Nurdinov N, Erdem S, Ozpolat B. Flavopiridol Suppresses Cell Proliferation and Migration and Induces Apoptotic Cell Death by Inhibiting Oncogenic FOXM1 Signaling in IDH Wild-Type and IDH-Mutant GBM Cells. Mol Neurobiol 2024; 61:1061-1079. [PMID: 37676393 DOI: 10.1007/s12035-023-03609-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/22/2023] [Indexed: 09/08/2023]
Abstract
Glioblastoma multiforme (GBM) remains one of the most challenging solid cancers to treat due to its highly aggressive and drug-resistant nature. Flavopiridol is synthetic flavone that was recently approved by the FDA for the treatment of acute myeloid leukemia. Flavopiridol exhibits antiproliferative activity in several solid cancer cells and currently evaluated in clinical trials in several solid and hematological cancers. In this study, we investigated the molecular mechanisms underlying antiproliferative effects of flavopiridol in GBM cell lines with wild-type and mutant encoding isocitrate dehydrogenase 1 (IDH1). We found that flavopiridol inhibits proliferation, colony formation, and migration and induces apoptosis in IDH1 wild-type and IDH-mutant cells through inhibition of FOXM1 oncogenic signaling. Furthermore, flavopiridol treatment also inhibits of NF-KB, mediators unfolded protein response (UPR), including, GRP78, PERK and IRE1α, and DNA repair enzyme PARP, which have been shown to be potential therapeutic targets by downregulating FOXM1 in GBM cells. Our findings suggest for the first time that flavopiridol suppresses proliferation, survival, and migration and induces apoptosis in IDH1 wild-type and IDH1-mutant GBM cells by targeting FOXM1 oncogenic signaling which also regulates NF-KB, PARP, and UPR response in GBM cells. Flavopiridol may be a potential novel therapeutic strategy in the treatment of patients IDH1 wild-type and IDH1-mutant GBM.
Collapse
Affiliation(s)
- Ahsen Guler
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
- Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
| | - Zuhal Hamurcu
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Turkey.
- Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey.
| | - Halil Ulutabanca
- Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
- Department of Neurosurgery, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Venhar Cınar
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
- Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
| | - Nursultan Nurdinov
- Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
- Faculties of Medicine and Dentistry, Ahmet Yesevi University, Turkestan, Kazakhstan
| | - Serife Erdem
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, Turkey
- Betül-Ziya Eren Genome and Stem Cell Center, Erciyes University, Kayseri, Turkey
| | - Bulent Ozpolat
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA.
- Methodist Neil Cancer Center, Houston, TX, USA.
| |
Collapse
|
14
|
Kosianova А, Pak O, Bryukhovetskiy I. Regulation of cancer stem cells and immunotherapy of glioblastoma (Review). Biomed Rep 2024; 20:24. [PMID: 38170016 PMCID: PMC10758921 DOI: 10.3892/br.2023.1712] [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: 09/06/2023] [Accepted: 11/24/2023] [Indexed: 01/05/2024] Open
Abstract
Glioblastoma (GB) is one of the most adverse diagnoses in oncology. Complex current treatment results in a median survival of 15 months. Resistance to treatment is associated with the presence of cancer stem cells (CSCs). The present review aimed to analyze the mechanisms of CSC plasticity, showing the particular role of β-catenin in regulating vital functions of CSCs, and to describe the molecular mechanisms of Wnt-independent increase of β-catenin levels, which is influenced by the local microenvironment of CSCs. The present review also analyzed the reasons for the low effectiveness of using medication in the regulation of CSCs, and proposed the development of immunotherapy scenarios with tumor cell vaccines, containing heterogenous cancer cells able of producing a multidirectional antineoplastic immune response. Additionally, the possibility of managing lymphopenia by transplanting hematopoietic stem cells from a healthy sibling and using clofazimine or other repurposed drugs that reduce β-catenin concentration in CSCs was discussed in the present study.
Collapse
Affiliation(s)
- Аleksandra Kosianova
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| | - Oleg Pak
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| | - Igor Bryukhovetskiy
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| |
Collapse
|
15
|
Rybarczyk A, Majchrzak-Celińska A, Krajka-Kuźniak V. Targeting Nrf2 Signaling Pathway in Cancer Prevention and Treatment: The Role of Cannabis Compounds. Antioxidants (Basel) 2023; 12:2052. [PMID: 38136172 PMCID: PMC10740807 DOI: 10.3390/antiox12122052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/24/2023] [Accepted: 11/25/2023] [Indexed: 12/24/2023] Open
Abstract
The development and progression of cancer are associated with the dysregulation of multiple pathways involved in cell proliferation and survival, as well as dysfunction in redox balance, immune response, and inflammation. The master antioxidant pathway, known as the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, regulates the cellular defense against oxidative stress and inflammation, making it a promising cancer prevention and treatment target. Cannabinoids have demonstrated anti-tumor and anti-inflammatory properties, affecting signaling pathways, including Nrf2. Increased oxidative stress following exposure to anti-cancer therapy prompts cancer cells to activate antioxidant mechanisms. This indicates the dual effect of Nrf2 in cancer cells-influencing proliferation and apoptotic processes and protecting against the toxicity of anti-cancer therapy. Therefore, understanding the complex role of cannabinoids in modulating Nrf2 might shed light on its potential implementation as an anti-cancer support. In this review, we aim to highlight the impact of cannabinoids on Nrf2-related factors, with a focus on cancer prevention and treatment. Additionally, we have presented the results of several research studies that combined cannabidiol (CBD) with other compounds targeting Nrf2. Further studies should be directed toward exploring the anti-inflammatory effects of cannabinoids in the context of cancer prevention and therapy.
Collapse
Affiliation(s)
| | | | - Violetta Krajka-Kuźniak
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3, 60-806 Poznań, Poland; (A.R.); (A.M.-C.)
| |
Collapse
|
16
|
Kwon IS, Hwang YN, Park JH, Na HH, Kwon TH, Park JS, Kim KC. Metallothionein Family Proteins as Regulators of Zinc Ions Synergistically Enhance the Anticancer Effect of Cannabidiol in Human Colorectal Cancer Cells. Int J Mol Sci 2023; 24:16621. [PMID: 38068944 PMCID: PMC10705991 DOI: 10.3390/ijms242316621] [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: 10/27/2023] [Revised: 11/17/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Cannabidiol (CBD) is a chemical obtained from Cannabis sativa; it has therapeutic effects on anxiety and cognition and anti-inflammatory properties. Although pharmacological applications of CBD in many types of tumors have recently been reported, the mechanism of action of CBD is not yet fully understood. In this study, we perform an mRNA-seq analysis to identify the target genes of CBD after determining the cytotoxic concentrations of CBD using an MTT assay. CBD treatment regulated the expression of genes related to DNA repair and cell division, with metallothionein (MT) family genes being identified as having highly increased expression levels induced by CBD. It was also found that the expression levels of MT family genes were decreased in colorectal cancer tissues compared to those in normal tissues, indicating that the downregulation of MT family genes might be highly associated with colorectal tumor progression. A qPCR experiment revealed that the expression levels of MT family genes were increased by CBD. Moreover, MT family genes were regulated by CBD or crude extract but not by other cannabinoids, suggesting that the expression of MT family genes was specifically induced by CBD. A synergistic effect between CBD and MT gene transfection or zinc ion treatment was found. In conclusion, MT family genes as novel target genes could synergistically increase the anticancer activity of CBD by regulating the zinc ions in human colorectal cancer cells.
Collapse
Affiliation(s)
- In-Seo Kwon
- Department of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon 24341, Kangwon, Republic of Korea; (I.-S.K.); (Y.-N.H.); (J.-H.P.); (H.-H.N.)
| | - Yu-Na Hwang
- Department of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon 24341, Kangwon, Republic of Korea; (I.-S.K.); (Y.-N.H.); (J.-H.P.); (H.-H.N.)
| | - Ju-Hee Park
- Department of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon 24341, Kangwon, Republic of Korea; (I.-S.K.); (Y.-N.H.); (J.-H.P.); (H.-H.N.)
| | - Han-Heom Na
- Department of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon 24341, Kangwon, Republic of Korea; (I.-S.K.); (Y.-N.H.); (J.-H.P.); (H.-H.N.)
- Kangwon Center for System Imaging, Chuncheon 24341, Kangwon, Republic of Korea
| | - Tae-Hyung Kwon
- Chuncheon Bioindustry Foundation, Chuncheon 24232, Kangwon, Republic of Korea;
| | - Jin-Sung Park
- Korean Pharmacopuncture Institute, Seoul 07525, Republic of Korea;
| | - Keun-Cheol Kim
- Department of Biological Sciences, College of Natural Sciences, Kangwon National University, Chuncheon 24341, Kangwon, Republic of Korea; (I.-S.K.); (Y.-N.H.); (J.-H.P.); (H.-H.N.)
- Kangwon Center for System Imaging, Chuncheon 24341, Kangwon, Republic of Korea
| |
Collapse
|
17
|
Li J, Gu T, Hu S, Jin B. Anti-proliferative effect of Cannabidiol in Prostate cancer cell PC3 is mediated by apoptotic cell death, NFκB activation, increased oxidative stress, and lower reduced glutathione status. PLoS One 2023; 18:e0286758. [PMID: 37796968 PMCID: PMC10553363 DOI: 10.1371/journal.pone.0286758] [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] [Received: 01/07/2023] [Accepted: 05/23/2023] [Indexed: 10/07/2023] Open
Abstract
Prostate cancer is the second most frequent cancer diagnosed in men in the world today. Almost all prostate cancers are adenocarcinomas and develop from gland cells. We used the PC3 prostate cancer cell line, which is well studied and derived from a bone metastasis of a grade IV prostatic adenocarcinoma. Cannabidiol (CBD), a major non-psychoactive constituent of cannabis, is a cannabinoid with anti-tumor properties but its effects on prostate cancer cells are not studied in detail. Here, we found cannabidiol decreased prostate cancer cell (PC3) viability up to 37.25% and induced apoptotic cell death in a time and dose-dependent manner. We found that CBD activated the caspases 3/7 pathways and increased DNA fragmentation. Furthermore, we observed an increase of pro-apoptotic genes Bax, an increased level of reactive oxygen species, lower reduced glutathione level, and altered mitochondrial potential in response to CBD treatment leading to lower cellular ATP. Overall, our results suggest that CBD may be effective against prostate cancer cells.
Collapse
Affiliation(s)
- Jie Li
- Department of Urology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
- Department of Urology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Tengfei Gu
- Department of Urology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Shengping Hu
- Department of Urology, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical College, Lishui, China
| | - Baiye Jin
- Department of Urology, Zhejiang University School of Medicine First Affiliated Hospital, Hangzhou, China
- Zhejiang Engineering Research Center for Urinary Bladder Carcinoma Innovation Diagnosis and Treatment, Hangzhou, China
| |
Collapse
|
18
|
Khodadadi H, Salles ÉL, Alptekin A, Mehrabian D, Rutkowski M, Arbab AS, Yeudall WA, Yu JC, Morgan JC, Hess DC, Vaibhav K, Dhandapani KM, Baban B. Inhalant Cannabidiol Inhibits Glioblastoma Progression Through Regulation of Tumor Microenvironment. Cannabis Cannabinoid Res 2023; 8:824-834. [PMID: 34918964 PMCID: PMC10589502 DOI: 10.1089/can.2021.0098] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Introduction: Glioblastoma (GBM) is the most common invasive brain tumor composed of diverse cell types with poor prognosis. The highly complex tumor microenvironment (TME) and its interaction with tumor cells play important roles in the development, progression, and durability of GBM. Angiogenic and immune factors are two major components of TME of GBM; their interplay is a major determinant of tumor vascularization, immune profile, as well as immune unresponsiveness of GBM. Given the ineffectiveness of current standard therapies (surgery, radiotherapy, and concomitant chemotherapy) in managing patients with GBM, it is necessary to develop new ways of treating these lethal brain tumors. Targeting TME, altering tumor ecosystem may be a viable therapeutic strategy with beneficial effects for patients in their fight against GBM. Materials and Methods: Given the potential therapeutic effects of cannabidiol (CBD) in a wide spectrum of diseases, including malignancies, we tested, for the first time, whether inhalant CBD can inhibit GBM tumor growth using a well-established orthotopic murine model. Optical imaging, histology, immunohistochemistry, and flow cytometry were employed to describe the outcomes such as tumor progression, cancer cell signaling pathways, and the TME. Results: Our findings showed that inhalation of CBD was able to not only limit the tumor growth but also to alter the dynamics of TME by repressing P-selectin, apelin, and interleukin (IL)-8, as well as blocking a key immune checkpoint-indoleamine 2,3-dioxygenase (IDO). In addition, CBD enhanced the cluster of differentiation (CD) 103 expression, indicating improved antigen presentation, promoted CD8 immune responses, and reduced innate Lymphoid Cells within the tumor. Conclusion: Overall, our novel findings support the possible therapeutic role of inhaled CBD as an effective, relatively safe, and easy to administer treatment adjunct for GBM with significant impacts on the cellular and molecular signaling of TME, warranting further research.
Collapse
Affiliation(s)
- Hesam Khodadadi
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
- Center for Excellence in Research, Scholarship and Innovation, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Évila Lopes Salles
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
- Center for Excellence in Research, Scholarship and Innovation, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Ahmet Alptekin
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Daniel Mehrabian
- Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Martin Rutkowski
- Department of Neurosurgery and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Ali S. Arbab
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - W. Andrew Yeudall
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
- Center for Excellence in Research, Scholarship and Innovation, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Jack C. Yu
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - John C. Morgan
- Parkinson's Foundation Center of Excellence, Movement Disorders, Program, Department of Neurology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - David C. Hess
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Kumar Vaibhav
- Department of Neurosurgery and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Krishnan M. Dhandapani
- Department of Neurosurgery and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
- Center for Excellence in Research, Scholarship and Innovation, Dental College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
19
|
Bai W, Cui F, Wang Z, Gu X, Fang X, Zhou L, Guo S. Receptor-interacting protein kinase 1 (RIPK1) regulates cervical cancer cells via NF-κB-TNF-α pathway: An in vitro study. Transl Oncol 2023; 36:101748. [PMID: 37516007 PMCID: PMC10410169 DOI: 10.1016/j.tranon.2023.101748] [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: 05/13/2023] [Revised: 07/02/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023] Open
Abstract
INTRODUCTION Cervical cancer (CC) is associated with high morbidity and mortality rates in women. Members of the receptor-interacting protein kinase (RIPK) family are important regulators of inflammation and cell death. However, the characteristics, molecular functions, and expression mechanisms of RIPK1 in CC remain unclear. MATERIAL AND METHODS To determine whether RIPK1 can be used for targeted therapy of CC, we assessed the clinical importance, biological function, and potential impact of RIPK1 in CC in 50 patients with CC. We utilized immunohistochemical staining, transfection, western blotting, cell counting kit-8 assay, colony formation assay, and wound healing assays among others, to elucidate the role of RIPK1 in CC. RESULTS RIPK1 expression was higher in tumor tissues than in paracancerous tissues. Poor prognosis of CC was linked to RIPK1 upregulation. Furthermore, silencing RIPK1 significantly inhibited the proliferation, migration, and invasion of CC cells in vitro. We also established that RIPK1 increased cell migration, invasion, and multiplication by regulating nuclear factor kappa-B (NF-κB) and tumor necrosis factor (TNF). DISCUSSION RIPK1 activates NF-κB and regulates TNF release to enhance the proliferation and spread of CC cells while suppressing their apoptosis. Therefore, RIPK1 plays a key role in the formation and progression of CC and is a potential target for CC treatment.
Collapse
Affiliation(s)
- Wenqi Bai
- Departments of Oncology Gynecology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu 233004, China
| | - Fengjie Cui
- Departments of Oncology Gynecology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu 233004, China
| | - Zihan Wang
- Department of Oncology Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Xianhua Gu
- Departments of Oncology Gynecology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu 233004, China
| | - Xiaojing Fang
- Departments of Oncology Gynecology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu 233004, China
| | - Li Zhou
- Departments of Oncology Gynecology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu 233004, China
| | - Suyang Guo
- Departments of Oncology Gynecology, The First Affiliated Hospital of Bengbu Medical College, No. 287 Changhuai Road, Bengbu 233004, China.
| |
Collapse
|
20
|
Ni B, Liu Y, Dai M, Zhao J, Liang Y, Yang X, Han B, Jiang M. The role of cannabidiol in aging. Biomed Pharmacother 2023; 165:115074. [PMID: 37418976 DOI: 10.1016/j.biopha.2023.115074] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 07/09/2023] Open
Abstract
Aging is usually considered a key risk factor associated with multiple diseases, such as neurodegenerative diseases, cardiovascular diseases and cancer. Furthermore, the burden of age-related diseases has become a global challenge. It is of great significance to search for drugs to extend lifespan and healthspan. Cannabidiol (CBD), a natural nontoxic phytocannabinoid, has been regarded as a potential candidate drug for antiaging. An increasing number of studies have suggested that CBD could benefit healthy longevity. Herein, we summarized the effect of CBD on aging and analyzed the possible mechanism. All these conclusions may provide a perspective for further study of CBD on aging.
Collapse
Affiliation(s)
- Beibei Ni
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yanying Liu
- Department of Basic Medical, Qingdao Huanghai University, Qingdao 266427, China
| | - Meng Dai
- Department of Emergency Internal Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Jun Zhao
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yu Liang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Xue Yang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Bing Han
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Man Jiang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| |
Collapse
|
21
|
Zhao P, Ma G, Ma L. miR-181a-5p targets DDX3X to inhibit the progression of osteoarthritis via NF-ΚB signaling pathway. J Orthop Surg Res 2023; 18:606. [PMID: 37587519 PMCID: PMC10433630 DOI: 10.1186/s13018-023-04073-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 08/03/2023] [Indexed: 08/18/2023] Open
Abstract
Osteoarthritis (OA) is the most common age-related joint disease, characterized by chronic inflammation, progressive articular cartilage destruction and subchondral osteosclerosis. More and more evidence showed that microRNAs (miRNAs) play a key role in various diseases, but the specific mechanism of miRNAs in OA is not clear. The purpose of this study was to investigate the expression level and role of miR-181a-5p in OA and its related mechanism. Here we identified the key gene DEAD-box RNA helicase 3X (DDX3X) in the OA dataset by bioinformatics analysis. At the same time, miRNAs targeting DDX3X were screened, and miR-181a-5p was selected as the next research object. Then we used different concentrations of interleukin-1 beta (IL-1β)-induced in vitro model of arthritis, and found that IL-1β can stimulate cells to release nitric oxide. The expression levels of miR-181a-5p and DDX3X in mouse chondrocyte cell line ATDC5 induced by IL-1β at a concentration of 10ug/mL were detected by reverse transcription-quantitative polymerase chain reaction (RT-qPCR). IL-1β induced a decrease in the expression of miR-181a-5p and an increase in the expression of DDX3X in ATDC5 cells. mimic miR-181a-5p or inhibitor miR-181a-5p were transfected into ATDC5 cells, and the levels of inflammatory mediators in the cells were detected by enzyme-linked immunosorbent assay, and the results showed that miR-181a-5p could reduce the release of tumor necrosis factor-α, IL-1β, IL-6 and inducible nitric oxide nitric oxide synthase in a cellular model of arthritis. Luciferase reporter assays confirmed that the miR-181a-5p binding site was in the DDX3X gene 3'-untranslated region (3'-UTR), and DDX3X was negatively regulated by miR-181a-5p. Rescue assays confirmed that miR-181a-5p reduced the expression of DDX3X by targeting the 3'-UTR region of DDX3X, thereby reducing the release of inflammatory factors. Finally, in this paper, western blot was used to detect the mechanism of miR-181a-5p regulating OA. The results showed that interfering with the expression of miR-181a-5p could up-regulate the expression of DDX3X protein, increase the expression of nuclear factor- kappaB (NF-κB) related proteins, and reduce the inflammatory response of OA, thereby increasing the secretion of the matrix proteinases MMP-3 and MMP-13. Taken together, the results of the study suggested that miR-181a-5p may be a promising therapeutic target for the treatment of human OA.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Rheumatology Immunology, 3201 Hospital, 783 Tianhan Avenue, Hantai District, Hanzhong, 723000, China.
| | - Guobin Ma
- Department of Rheumatology Immunology, 3201 Hospital, 783 Tianhan Avenue, Hantai District, Hanzhong, 723000, China
| | - Lintong Ma
- Department of Hematology, 3201 Hospital, Hanzhong, China
| |
Collapse
|
22
|
Li W, Xu X. Advances in mitophagy and mitochondrial apoptosis pathway-related drugs in glioblastoma treatment. Front Pharmacol 2023; 14:1211719. [PMID: 37456742 PMCID: PMC10347406 DOI: 10.3389/fphar.2023.1211719] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/23/2023] [Indexed: 07/18/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant tumor of the central nervous system (CNS). It is a leading cause of death among patients with intracranial malignant tumors. GBM exhibits intra- and inter-tumor heterogeneity, leading to drug resistance and eventual tumor recurrence. Conventional treatments for GBM include maximum surgical resection of glioma tissue, temozolomide administration, and radiotherapy, but these methods do not effectively halt cancer progression. Therefore, development of novel methods for the treatment of GBM and identification of new therapeutic targets are urgently required. In recent years, studies have shown that drugs related to mitophagy and mitochondrial apoptosis pathways can promote the death of glioblastoma cells by inducing mitochondrial damage, impairing adenosine triphosphate (ATP) synthesis, and depleting large amounts of ATP. Some studies have also shown that modern nano-drug delivery technology targeting mitochondria can achieve better drug release and deeper tissue penetration, suggesting that mitochondria could be a new target for intervention and therapy. The combination of drugs targeting mitochondrial apoptosis and autophagy pathways with nanotechnology is a promising novel approach for treating GBM.This article reviews the current status of drug therapy for GBM, drugs targeting mitophagy and mitochondrial apoptosis pathways, the potential of mitochondria as a new target for GBM treatment, the latest developments pertaining to GBM treatment, and promising directions for future research.
Collapse
|
23
|
Yue M, Guo T, Nie DY, Zhu YX, Lin M. Advances of nanotechnology applied to cancer stem cells. World J Stem Cells 2023; 15:514-529. [PMID: 37424953 PMCID: PMC10324502 DOI: 10.4252/wjsc.v15.i6.514] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/01/2023] [Accepted: 04/18/2023] [Indexed: 06/26/2023] Open
Abstract
Cancer stem cells (CSCs) are a small proportion of the cells that exist in cancer tissues. They are considered to be the culprit of tumor genesis, development, drug resistance, metastasis and recurrence because of their self-renewal, proliferation, and differentiation potential. The elimination of CSCs is thus the key to cure cancer, and targeting CSCs provides a new method for tumor treatment. Due to the advantages of controlled sustained release, targeting and high biocompatibility, a variety of nanomaterials are used in the diagnosis and treatments targeting CSCs and promote the recognition and removal of tumor cells and CSCs. This article mainly reviews the research progress of nanotechnology in sorting CSCs and nanodrug delivery systems targeting CSCs. Furthermore, we identify the problems and future research directions of nanotechnology in CSC therapy. We hope that this review will provide guidance for the design of nanotechnology as a drug carrier so that it can be used in clinic for cancer therapy as soon as possible.
Collapse
Affiliation(s)
- Miao Yue
- Clinical Laboratory, Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu Province, China
| | - Ting Guo
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu Province, China
| | - Deng-Yun Nie
- Clinical Laboratory, Nanjing University of Chinese Medicine, Taizhou 225300, Jiangsu Province, China
| | - Yin-Xing Zhu
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu Province, China
| | - Mei Lin
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, Jiangsu Province, China.
| |
Collapse
|
24
|
Shi P, Xu J, Cui H. The Recent Research Progress of NF-κB Signaling on the Proliferation, Migration, Invasion, Immune Escape and Drug Resistance of Glioblastoma. Int J Mol Sci 2023; 24:10337. [PMID: 37373484 PMCID: PMC10298967 DOI: 10.3390/ijms241210337] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/09/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and invasive primary central nervous system tumor in humans, accounting for approximately 45-50% of all primary brain tumors. How to conduct early diagnosis, targeted intervention, and prognostic evaluation of GBM, in order to improve the survival rate of glioblastoma patients, has always been an urgent clinical problem to be solved. Therefore, a deeper understanding of the molecular mechanisms underlying the occurrence and development of GBM is also needed. Like many other cancers, NF-κB signaling plays a crucial role in tumor growth and therapeutic resistance in GBM. However, the molecular mechanism underlying the high activity of NF-κB in GBM remains to be elucidated. This review aims to identify and summarize the NF-κB signaling involved in the recent pathogenesis of GBM, as well as basic therapy for GBM via NF-κB signaling.
Collapse
Affiliation(s)
- Pengfei Shi
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China; (P.S.); (J.X.)
- Jinfeng Laboratory, Chongqing 401329, China
| | - Jie Xu
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China; (P.S.); (J.X.)
- Jinfeng Laboratory, Chongqing 401329, China
| | - Hongjuan Cui
- Cancer Center, Medical Research Institute, Southwest University, Chongqing 400716, China; (P.S.); (J.X.)
- Jinfeng Laboratory, Chongqing 401329, China
- State Key Laboratory of Resource Insects, Southwest University, Chongqing 400716, China
| |
Collapse
|
25
|
Xu F, Wang J, Zhen S, Duan Y, Li Q, Liu L. C1ql4 regulates breast cancer cell stemness and epithelial-mesenchymal transition through PI3K/AKT/NF-κB signaling pathway. Front Oncol 2023; 13:1192482. [PMID: 37324011 PMCID: PMC10265994 DOI: 10.3389/fonc.2023.1192482] [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: 03/23/2023] [Accepted: 05/12/2023] [Indexed: 06/17/2023] Open
Abstract
Background The stemness characteristic of breast cancer (BC) is a crucial factor underlying cancer recurrence and metastasis after operative therapy and chemoradiotherapy. Understanding the potential mechanism of breast cancer stem cells (BCSCs) may ameliorate the prognosis of patients. Methods We collected clinical specimens of BC patients for staining and statistical analysis to verify the expression status and clinical significance of complement C1q-like 4 (C1ql4). Western blot and qRT-PCR were employed to detect the expression of molecules. Flow cytometry was used to examine cell cycle, cell apoptosis and the portion of BCSCs. Wound healing and Transwell assays were used to detect cell metastasis. The effect of C1ql4 on breast cancer progression in vivo was examined in a nude mouse tumor bearing model. Results Our clinical analysis showed that C1ql4 was highly expressed in BC tissues and cell lines, and the high expression of C1ql4 was significantly corelated with the malignancy of BC patients. Moreover, we also found that C1ql4 was overexpressed in BCSCs. C1ql4 knockdown suppressed the BCSC and EMT properties, promoted cell cycle progression, enhanced BC cell apoptosis, and inhibited cell migration and invasion, whereas the C1ql4 overexpression exhibited the opposite effects. Mechanistically, C1ql4 promoted the activation and nuclear location of NF-κB and the expression of downstream factors TNF-α and IL-1β. Moreover, inhibition of PI3K/AKT signaling suppressed the C1ql4-induced stemness and EMT. Conclusions Our findings suggest that C1ql4 promotes the BC cell stemness and EMT via modulating the PI3K/AKT/NF-κB signaling, and provides a promising target for BC treatment.
Collapse
Affiliation(s)
- Fan Xu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Departments of Oncology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Jiali Wang
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shuman Zhen
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yuqing Duan
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Qingshan Li
- Departments of Oncology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Lihua Liu
- Department of Tumor Immunotherapy, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
26
|
Antitumor Potential of Antiepileptic Drugs in Human Glioblastoma: Pharmacological Targets and Clinical Benefits. Biomedicines 2023; 11:biomedicines11020582. [PMID: 36831117 PMCID: PMC9953000 DOI: 10.3390/biomedicines11020582] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/07/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Glioblastoma (GBM) is characterized by fast-growing cells, genetic and phenotypic heterogeneity, and radio-chemo-therapy resistance, contributing to its dismal prognosis. Various medical comorbidities are associated with the natural history of GBM. The most disabling and greatly affecting patients' quality of life are neurodegeneration, cognitive impairment, and GBM-related epilepsy (GRE). Hallmarks of GBM include molecular intrinsic mediators and pathways, but emerging evidence supports the key role of non-malignant cells within the tumor microenvironment in GBM aggressive behavior. In this context, hyper-excitability of neurons, mediated by glutamatergic and GABAergic imbalance, contributing to GBM growth strengthens the cancer-nervous system crosstalk. Pathogenic mechanisms, clinical features, and pharmacological management of GRE with antiepileptic drugs (AEDs) and their interactions are poorly explored, yet it is a potentially promising field of research in cancer neuroscience. The present review summarizes emerging cooperative mechanisms in oncogenesis and epileptogenesis, focusing on the neuron-to-glioma interface. The main effects and efficacy of selected AEDs used in the management of GRE are discussed in this paper, as well as their potential beneficial activity as antitumor treatment. Overall, although still many unclear processes overlapping in GBM growth and seizure onset need to be elucidated, this review focuses on the intriguing targeting of GBM-neuron mutual interactions to improve the outcome of the so challenging to treat GBM.
Collapse
|
27
|
Induction of apoptosis in glioma cells by lycorine via reactive oxygen species generation and regulation of NF-κB pathways. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1247-1255. [PMID: 36715733 DOI: 10.1007/s00210-023-02384-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 01/02/2023] [Indexed: 01/31/2023]
Abstract
Glioma is an extremely aggressive primary brain tumor, which is highly resistant to chemotherapy, presenting a therapeutic challenge. Here, we explored the anti-glioma effects and the underlying mechanism of lycorine, an isoquinoline alkaloid isolated from lycoris on glioma cells. We found that lycorine could dose dependently inhibit C6 glioma cell growth and induce cell apoptosis and intracellular reactive oxygen species (ROS) production. The half-maximal inhibitory concentration (IC50) values of lycorine on C6 glioma cells at 48 h was 2.85 μM. Meanwhile, lycorine treatment caused dysfunction of the NF-κB signal, as demonstrated by the up-regulation of NF-κB inhibitor protein IκB and the downregulation of the NF-κB phosphorylation protein p-p65. The addition of NF-κB inhibitor SC75741 further confirmed the importance of the NF-κB pathway in lycorine-induced cell-growth inhibition. Moreover, lycorine might act synergically with temozolomide (TMZ) to reduce drug resistance by blocking the NF-κB pathway. Our study suggested that lycorine exerts an anti-glioma effect by inducing ROS production and inhibiting NF-κB, which validated that lycorine may be a potential candidate for glioma treatment alone or in combination with TMZ.
Collapse
|
28
|
Atalay Ekiner S, Gęgotek A, Skrzydlewska E. The molecular activity of cannabidiol in the regulation of Nrf2 system interacting with NF-κB pathway under oxidative stress. Redox Biol 2022; 57:102489. [PMID: 36198205 PMCID: PMC9535304 DOI: 10.1016/j.redox.2022.102489] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/11/2022] [Accepted: 09/22/2022] [Indexed: 11/30/2022] Open
Abstract
Cannabidiol (CBD), the major non-psychoactive phytocannabinoid of Cannabis sativa L., is one of the most studied compounds in pharmacotherapeutic approaches to treat oxidative stress-related diseases such as cardiovascular, metabolic, neurodegenerative, and neoplastic diseases. The literature data to date indicate the possibility of both antioxidant and pro-oxidative effects of CBD. Thus, the mechanism of action of this natural compound in the regulation of nuclear factor 2 associated with erythroid 2 (Nrf2), which plays the role of the main cytoprotective regulator of redox balance and inflammation under oxidative stress conditions, seems to be particularly important. Moreover, Nrf2 is strongly correlated with the cellular neoplastic profile and malignancy, which in turn is critical in determining the cellular response induced by CBD under pathophysiological conditions. This paper summarizes the CBD-mediated pathways of regulation of the Nrf2 system by altering the expression and modification of both proteins directly involved in Nrf2 transcriptional activity and proteins involved in the relationship between Nrf2 and the nuclear factor kappa B (NF-κB) which is another redox-sensitive transcription factor.
Collapse
Affiliation(s)
- Sinemyiz Atalay Ekiner
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland.
| | - Agnieszka Gęgotek
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland.
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland.
| |
Collapse
|
29
|
Zhu Z, Fang C, Xu H, Yuan L, Du Y, Ni Y, Xu Y, Shao A, Zhang A, Lou M. Anoikis resistance in diffuse glioma: The potential therapeutic targets in the future. Front Oncol 2022; 12:976557. [PMID: 36046036 PMCID: PMC9423707 DOI: 10.3389/fonc.2022.976557] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 07/25/2022] [Indexed: 11/28/2022] Open
Abstract
Glioma is the most common malignant intracranial tumor and exhibits diffuse metastasis and a high recurrence rate. The invasive property of glioma results from cell detachment. Anoikis is a special form of apoptosis that is activated upon cell detachment. Resistance to anoikis has proven to be a protumor factor. Therefore, it is suggested that anoikis resistance commonly occurs in glioma and promotes diffuse invasion. Several factors, such as integrin, E-cadherin, EGFR, IGFR, Trk, TGF-β, the Hippo pathway, NF-κB, eEF-2 kinase, MOB2, hypoxia, acidosis, ROS, Hsp and protective autophagy, have been shown to induce anoikis resistance in glioma. In our present review, we aim to summarize the underlying mechanism of resistance and the therapeutic potential of these molecules.
Collapse
Affiliation(s)
- Zhengyang Zhu
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaoyou Fang
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Houshi Xu
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ling Yuan
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yichao Du
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yunjia Ni
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuanzhi Xu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Anke Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
- Department of Neurosurgery, Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Meiqing Lou
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
30
|
Comparing Tumor Cell Invasion and Myeloid Cell Composition in Compatible Primary and Relapsing Glioblastoma. Cancers (Basel) 2021; 13:cancers13143636. [PMID: 34298846 PMCID: PMC8303884 DOI: 10.3390/cancers13143636] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary We established a new minimally invasive mouse model for GBM relapse. For this, we utilized orthotopical implantation of HSVTK-transduced GBM cells and pharmacological treatment with GCV. In addition, we implanted patient-derived GBM cells of primary or recurrent tumors. We found that recurrent GBM were more aggressively invasive than primary GBM. Moreover, the recurring tumors had a higher ratio of monocyte-derived macrophages among the entire population of tumor associated myeloid cells. This shift in the composition of tumor-associated immune cells appeared to be independent from cell-death signaling or surgical intervention. This model provides the means to investigate the entire process of tumor relapse and test standard as well as experimental therapeutic strategies for relapsing GBM under defined conditions. Abstract Glioblastoma (GBM) recurrence after treatment is almost inevitable but addressing this issue with adequate preclinical models has remained challenging. Here, we introduce a GBM mouse model allowing non-invasive and scalable de-bulking of a tumor mass located deeply in the brain, which can be combined with conventional therapeutic approaches. Strong reduction of the GBM volume is achieved after pharmacologically inducing a tumor-specific cell death mechanism. This is followed by GBM re-growth over a predictable timeframe. Pharmacological de-bulking followed by tumor relapse was accomplished with an orthotopic mouse glioma model. Relapsing experimental tumors recapitulated pathological features often observed in recurrent human GBM, like increased invasiveness or altered immune cell composition. Orthotopic implantation of GBM cells originating from biopsies of one patient at initial or follow-up treatment reproduced these findings. Interestingly, relapsing GBM of both models contained a much higher ratio of monocyte-derived macrophages (MDM) versus microglia than primary GBM. This was not altered when combining pharmacological de-bulking with invasive surgery. We interpret that factors released from viable primary GBM cells preferentially attract microglia whereas relapsing tumors preponderantly release chemoattractants for MDM. All in all, this relapse model has the capacity to provide novel insights into clinically highly relevant aspects of GBM treatment.
Collapse
|