1
|
Singh A, Chauhan R, Rajput VD, Minkina T, Prasad R, Goel A. Exploring the insights of bioslurry-Nanoparticle amalgam for soil amelioration. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:58310-58323. [PMID: 39307866 DOI: 10.1007/s11356-024-35003-0] [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: 06/18/2024] [Accepted: 09/13/2024] [Indexed: 10/11/2024]
Abstract
In response to global agricultural challenges, this review examines the synergistic impact of bioslurry and biogenic nanoparticles on soil amelioration. Bioslurry, rich in N, P, K and beneficial microorganisms, combined with zinc oxide nanoparticles synthesized through eco-friendly methods, demonstrates remarkable soil improvement capabilities. Their synergistic effects include enhanced nutrient availability through increased soil enzymatic activities, improved soil structure via stable aggregate formation, stimulated microbial activity particularly beneficial groups, enhanced water retention due to increased organic matter and modified soil surface properties and reduced soil pH fluctuations. These mechanisms significantly impact soil physico-chemical properties including cation exchange capacity, electrical conductivity and nutrient dynamics. This review analyses these effects and their implications for sustainable agricultural practices, focusing on crop yield improvements, reduced chemical fertilizer dependence and enhanced plant stress tolerance. Knowledge gaps such as long-term nanoparticle accumulation effects and impacts on non-target organisms are identified. Future research directions include optimizing bioslurry-nanoparticle ratios for various soil types and developing "smart" nanoparticle-enabled biofertilizers with controlled release properties. This innovative approach contributes to environmentally friendly farming practices, potentially enhancing global food security and supporting sustainable agriculture transitions. The integration of bioslurry and biogenic nanoparticles presents a promising solution to soil degradation and agricultural sustainability challenges.
Collapse
Affiliation(s)
- Abhinav Singh
- Amity Institute of Microbial Technology, Amity University, Noida, 201313, India
| | - Ritika Chauhan
- Amity Institute of Microbial Technology, Amity University, Noida, 201313, India
| | - Vishnu D Rajput
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, 344090, Russia
| | - Tatiana Minkina
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-on-Don, 344090, Russia
| | - Ram Prasad
- Department of Botany, Mahatma Gandhi Central University, Motihari, 845801, Bihar, India
| | - Arti Goel
- Amity Institute of Microbial Technology, Amity University, Noida, 201313, India.
| |
Collapse
|
2
|
Chaudhary AA, Fareed M, Khan SUD, Alneghery LM, Aslam M, Alex A, Rizwanullah M. Exploring the therapeutic potential of lipid-based nanoparticles in the management of oral squamous cell carcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1223-1246. [PMID: 39465011 PMCID: PMC11502080 DOI: 10.37349/etat.2024.00272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a highly malignant and invasive tumor with significant mortality and morbidity. Current treatment modalities such as surgery, radiotherapy, and chemotherapy encounter significant limitations, such as poor targeting, systemic toxicity, and drug resistance. There is an urgent need for novel therapeutic strategies that offer targeted delivery, enhanced efficacy, and reduced side effects. The advent of lipid-based nanoparticles (LNPs) offers a promising tool for OSCC therapy, potentially overcoming the limitations of current therapeutic approaches. LNPs are composed of biodegradable and biocompatible lipids, which minimize the risk of toxicity and adverse effects. LNPs can encapsulate hydrophobic drugs, improving their solubility and stability in the biological environment, thereby enhancing their bioavailability. LNPs demonstrate significantly higher ability to encapsulate lipophilic drugs than other nanoparticle types. LNPs offer excellent storage stability, minimal drug leakage, and controlled drug release, making them highly effective nanoplatforms for the delivery of chemotherapeutic agents. Additionally, LNPs can be modified by complexing them with specific target ligands on their surface. This surface modification allows the active targeting of LNPs to the tumors in addition to the passive targeting mechanism. Furthermore, the PEGylation of LNPs improves their hydrophilicity and enhances their biological half-life by reducing clearance by the reticuloendothelial system. This review aims to discuss current treatment approaches and their limitations, as well as recent advancements in LNPs for better management of OSCC.
Collapse
Affiliation(s)
- Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohammad Fareed
- College of Medicine, AlMaarefa University, Diriyah, Riyadh 11597, Saudi Arabia
| | - Salah-Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Lina M Alneghery
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohammed Aslam
- Pharmacy Department, Tishk International University, Erbil 44001, Kurdistan Region, Iraq
| | - Arockia Alex
- Molecular and Nanobiotechnology Laboratory (MNBL), Department of Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| | - Md Rizwanullah
- Drug Delivery and Nanomedicine Unit, Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| |
Collapse
|
3
|
Qu Z, Luo J, Li Z, Yang R, Zhao J, Chen X, Yu S, Shu H. Advancements in strategies for overcoming the blood-brain barrier to deliver brain-targeted drugs. Front Aging Neurosci 2024; 16:1353003. [PMID: 39253614 PMCID: PMC11381257 DOI: 10.3389/fnagi.2024.1353003] [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: 12/09/2023] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
The blood-brain barrier is known to consist of a variety of cells and complex inter-cellular junctions that protect the vulnerable brain from neurotoxic compounds; however, it also complicates the pharmacological treatment of central nervous system disorders as most drugs are unable to penetrate the blood-brain barrier on the basis of their own structural properties. This dramatically diminished the therapeutic effect of the drug and compromised its biosafety. In response, a number of drugs are often delivered to brain lesions in invasive ways that bypass the obstruction of the blood-brain barrier, such as subdural administration, intrathecal administration, and convection-enhanced delivery. Nevertheless, these intrusive strategies introduce the risk of brain injury, limiting their clinical application. In recent years, the intensive development of nanomaterials science and the interdisciplinary convergence of medical engineering have brought light to the penetration of the blood-brain barrier for brain-targeted drugs. In this paper, we extensively discuss the limitations of the blood-brain barrier on drug delivery and non-invasive brain-targeted strategies such as nanomedicine and blood-brain barrier disruption. In the meantime, we analyze their strengths and limitations and provide outlooks on the further development of brain-targeted drug delivery systems.
Collapse
Affiliation(s)
- Zhichuang Qu
- Department of Neurosurgery, Meishan City People's Hospital, Meishan, China
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Juan Luo
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Zheng Li
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Rong Yang
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiaxi Zhao
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- Department of Neurosurgery, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xin Chen
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
| | - Sixun Yu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine of Southwest Jiaotong University, Chengdu, China
| | - Haifeng Shu
- Department of Neurosurgery, General Hospital of Western Theater Command, Chengdu, China
- College of Medicine of Southwest Jiaotong University, Chengdu, China
| |
Collapse
|
4
|
Xie W, Lu J, Chen Y, Wang X, Lu H, Li Q, Jin N, He J, Ou L, Ni J, Shen Y, Shao L. TCL1A-expressing B cells are critical for tertiary lymphoid structure formation and the prognosis of oral squamous cell carcinoma. J Transl Med 2024; 22:477. [PMID: 38764038 PMCID: PMC11103841 DOI: 10.1186/s12967-024-05292-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 05/11/2024] [Indexed: 05/21/2024] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC) is a malignant tumor with a poor prognosis. Traditional treatments have limited effectiveness. Regulation of the immune response represents a promising new approach for OSCC treatment. B cells are among the most abundant immune cells in OSCC. However, the role of B cells in OSCC treatment has not been fully elucidated. METHODS Single-cell RNA sequencing analysis of 13 tissues and 8 adjacent normal tissues from OSCC patients was performed to explore differences in B-cell gene expression between OSCC tissues and normal tissues. We further investigated the relationship between differentially expressed genes and the immune response to OSCC. We utilized tissue microarray data for 146 OSCC clinical samples and RNA sequencing data of 359 OSCC samples from The Cancer Genome Atlas (TCGA) to investigate the role of T-cell leukemia 1 A (TCL1A) in OSCC prognosis. Multiplex immunohistochemistry (mIHC) was employed to investigate the spatial distribution of TCL1A in OSCC tissues. We then investigated the effect of TCL1A on B-cell proliferation and trogocytosis. Finally, lentiviral transduction was performed to induce TCL1A overexpression in B lymphoblastoid cell lines (BLCLs) to verify the function of TCL1A. RESULTS Our findings revealed that TCL1A was predominantly expressed in B cells and was associated with a better prognosis in OSCC patients. Additionally, we found that TCL1A-expressing B cells are located at the periphery of lymphatic follicles and are associated with tertiary lymphoid structures (TLS) formation in OSCC. Mechanistically, upregulation of TCL1A promoted the trogocytosis of B cells on dendritic cells by mediating the upregulation of CR2, thereby improving antigen-presenting ability. Moreover, the upregulation of TCL1A expression promoted the proliferation of B cells. CONCLUSION This study revealed the role of B-cell TCL1A expression in TLS formation and its effect on OSCC prognosis. These findings highlight TCL1A as a novel target for OSCC immunotherapy.
Collapse
Affiliation(s)
- Wenqiang Xie
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Jinjin Lu
- Department of Periodontics, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, PR China
| | - Yichen Chen
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Xi Wang
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, 510055, PR China
| | - Huanzi Lu
- Guanghua School of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Stomatological Hospital, Sun Yat-Sen University, Guangzhou, 510055, PR China
| | - Qunxing Li
- Department of Stomatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, PR China
| | - Nianqiang Jin
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Jiankang He
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Lingling Ou
- Department of Periodontics, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, PR China
| | - Jia Ni
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China
| | - Yuqin Shen
- Department of Periodontics, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, 510182, Guangdong, PR China
| | - Longquan Shao
- Stomatological Hospital, Southern Medical University, Guangzhou, 510280, PR China.
| |
Collapse
|
5
|
Zhang J, Di Y, Zhang B, Li T, Li D, Zhang H. CDK1 and CCNA2 play important roles in oral squamous cell carcinoma. Medicine (Baltimore) 2024; 103:e37831. [PMID: 38640322 PMCID: PMC11029925 DOI: 10.1097/md.0000000000037831] [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: 02/22/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/21/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a malignant tumor that occurs in oral cavity and is dominated by squamous cells. The relationship between CDK1, CCNA2, and OSCC is still unclear. The OSCC datasets GSE74530 and GSE85195 configuration files were downloaded from the Gene Expression Omnibus (GEO) database and were derived from platforms GPL570 and GPL6480. Differentially expressed genes (DEGs) were screened. The weighted gene co-expression network analysis, functional enrichment analysis, gene set enrichment analysis, construction and analysis of protein-protein interaction (PPI) network, Comparative Toxicogenomics Database analysis were performed. Gene expression heatmap was drawn. TargetScan was used to screen miRNAs that regulate central DEGs. A total of 1756 DEGs were identified. According to Gene Ontology (GO) analysis, they were predominantly enriched in processes related to organic acid catabolic metabolism, centromeric, and chromosomal region condensation, and oxidoreductase activity. In Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, the DEGs were mainly concentrated in metabolic pathways, P53 signaling pathway, and PPAR signaling pathway. Weighted gene co-expression network analysis was performed with a soft-thresholding power set at 9, leading to the identification of 6 core genes (BUB1B, CCNB1, KIF20A, CCNA2, CDCA8, CDK1). The gene expression heatmap revealed that core genes (CDK1, CCNA2) were highly expressed in OSCC samples. Comparative Toxicogenomics Database analysis demonstrated associations between the 6 genes (BUB1B, CCNB1, KIF20A, CCNA2, CDCA8, CDK1) and oral tumors, precancerous lesions, inflammation, immune system disorders, and tongue tumors. The associated miRNAs for CDK1 gene were hsa-miR-203a-3p.2, while for CCNA2 gene, they were hsa-miR-6766-3p, hsa-miR-4782-3p, and hsa-miR-219a-5p. CDK1 and CCNA2 are highly expressed in OSCC. The higher the expression of CDK1 and CCNA2, the worse the prognosis.
Collapse
Affiliation(s)
- Junbo Zhang
- Department of Stomatology, Tangshan Gongren Hospital, Tangshan City, China
| | - Yongbin Di
- Department of Stomatology, The First Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Bohao Zhang
- Department of Otolaryngology and Head and Neck Surgery, The First Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Tianke Li
- Department of Stomatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Dan Li
- Department of Otolaryngology and Head and Neck Surgery, The First Hospital of Hebei Medical University, Shijiazhuang City, China
| | - Haolei Zhang
- Department of Otolaryngology and Head and Neck Surgery, The First Hospital of Hebei Medical University, Shijiazhuang City, China
| |
Collapse
|
6
|
Lan Z, Liu WJ, Yin WW, Yang SR, Cui H, Zou KL, Cheng GW, Chen H, Han YH, Rao L, Tian R, Li LL, Zhao YY, Yu GT. Biomimetic MDSCs membrane coated black phosphorus nanosheets system for photothermal therapy/photodynamic therapy synergized chemotherapy of cancer. J Nanobiotechnology 2024; 22:174. [PMID: 38609922 PMCID: PMC11015563 DOI: 10.1186/s12951-024-02417-4] [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: 12/01/2023] [Accepted: 03/18/2024] [Indexed: 04/14/2024] Open
Abstract
Photothermal therapy is favored by cancer researchers due to its advantages such as controllable initiation, direct killing and immune promotion. However, the low enrichment efficiency of photosensitizer in tumor site and the limited effect of single use limits the further development of photothermal therapy. Herein, a photo-responsive multifunctional nanosystem was designed for cancer therapy, in which myeloid-derived suppressor cell (MDSC) membrane vesicle encapsulated decitabine-loaded black phosphorous (BP) nanosheets (BP@ Decitabine @MDSCs, named BDM). The BDM demonstrated excellent biosafety and biochemical characteristics, providing a suitable microenvironment for cancer cell killing. First, the BDM achieves the ability to be highly enriched at tumor sites by inheriting the ability of MDSCs to actively target tumor microenvironment. And then, BP nanosheets achieves hyperthermia and induces mitochondrial damage by its photothermal and photodynamic properties, which enhancing anti-tumor immunity mediated by immunogenic cell death (ICD). Meanwhile, intra-tumoral release of decitabine induced G2/M cell cycle arrest, further promoting tumor cell apoptosis. In vivo, the BMD showed significant inhibition of tumor growth with down-regulation of PCNA expression and increased expression of high mobility group B1 (HMGB1), calreticulin (CRT) and caspase 3. Flow cytometry revealed significantly decreased infiltration of MDSCs and M2-macrophages along with an increased proportion of CD4+, CD8+ T cells as well as CD103+ DCs, suggesting a potentiated anti-tumor immune response. In summary, BDM realizes photothermal therapy/photodynamic therapy synergized chemotherapy for cancer.
Collapse
Affiliation(s)
- Zhou Lan
- Stomatological Hospital, School of Stomatology, Southern Medical University, No 366, Jiangnan Road, Haizhu Region, Guangzhou City, China
| | - Wei-Jia Liu
- Department of Oral Mucosal Diseases, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, 510182, China
| | - Wu-Wei Yin
- Stomatological Hospital, School of Stomatology, Southern Medical University, No 366, Jiangnan Road, Haizhu Region, Guangzhou City, China
| | - Sheng-Ren Yang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Hao Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, No 366, Jiangnan Road, Haizhu Region, Guangzhou City, China
| | - Ke-Long Zou
- Stomatological Hospital, School of Stomatology, Southern Medical University, No 366, Jiangnan Road, Haizhu Region, Guangzhou City, China
| | - Guo-Wang Cheng
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Hao Chen
- Stomatological Hospital, School of Stomatology, Southern Medical University, No 366, Jiangnan Road, Haizhu Region, Guangzhou City, China
| | - Yan-Hua Han
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Lang Rao
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, 518132, China
| | - Rui Tian
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Ling-Ling Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, No 101, Longmian Road, Jiangning Region, Nanjing, 211166, China.
| | - Yu-Yue Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, No 366, Jiangnan Road, Haizhu Region, Guangzhou City, China.
| | - Guang-Tao Yu
- Stomatological Hospital, School of Stomatology, Southern Medical University, No 366, Jiangnan Road, Haizhu Region, Guangzhou City, China.
| |
Collapse
|
7
|
Ali Alghamdi M, Haider M, Intagliata S, Pittalà V, Jagal J, Haider Y, Althaf N, Greish K. Lauric acid-based thermosensitive delivery system for the treatment of head and neck squamous cell carcinoma. J Drug Target 2024; 32:433-443. [PMID: 38385752 DOI: 10.1080/1061186x.2024.2323056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 02/16/2024] [Indexed: 02/23/2024]
Abstract
Traditional treatments for head and neck squamous cell carcinoma (HNSCC) such as surgery, radiation therapy, and chemotherapy, often have severe side effects. Local delivery of chemotherapeutic agents can be a promising approach to minimise systemic toxicity and improve efficacy. Lauric acid (LA), was explored as a novel injectable thermosensitive drug reservoir as a depot for sustained release of anticancer drugs to treat HNSCC. LA was characterised in terms of melting temperature and gelation time. The efficacy of LA-based drug formulations was tested in vitro in a HNSCC cell line and in vivo in a mouse model of HNSCC. LA was modified to have a melting point of 38.5 °C and a gelation time of 40 s at 37.5 °C, rendering it suitable for injection at body temperature. LA- based doxorubicin (DOXO) formulation showed slow release with a maximum of 18% release after 3 days. The in vitro study showed that LA enhanced the cytotoxic effect of DOXO. LA combined with DOXO prevented tumour progression and LA alone significantly reduced the original tumour volume compared to the untreated control group. These findings confirmed that LA can function as practical carrier for the local delivery of chemotherapeutics and provides a safe and simple strategy for the delivery of hydrophobic anticancer drugs and warrant further testing in clinical trials.
Collapse
Affiliation(s)
- Maha Ali Alghamdi
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
- Department of Molecular Medicine, Princess Al-Jawhara Centre for Molecular Medicine, School of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Mohamed Haider
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Valeria Pittalà
- Department of Molecular Medicine, Princess Al-Jawhara Centre for Molecular Medicine, School of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
- Department of Drug and Health Science, University of Catania, CT, Italy
| | - Jayalakshmi Jagal
- Research Institute of Medical & Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Youssef Haider
- College of Engineering, Boston University, Boston, MA, USA
| | - Nasneen Althaf
- Animal House Facility Unit, College of Medicine & Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | - Khaled Greish
- Department of Molecular Medicine, Princess Al-Jawhara Centre for Molecular Medicine, School of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| |
Collapse
|
8
|
Beniwal N, Verma A, Putta CL, Rengan AK. Recent Trends in Bio-nanomaterials and Non-invasive Combinatorial Approaches of Photothermal Therapy against Cancer. Nanotheranostics 2024; 8:219-238. [PMID: 38444743 PMCID: PMC10911972 DOI: 10.7150/ntno.91356] [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: 10/20/2023] [Accepted: 01/16/2024] [Indexed: 03/07/2024] Open
Abstract
In 2020, approximately 10 million deaths worldwide were attributed to cancer, making it the primary cause of death globally. Photothermal therapy (PTT) is one of the novel ways to treat and abolish cancer. PTT significantly impacts cancer theranostics compared to other therapies like surgery, chemotherapy, and radiotherapy due to its remarkable binding capability to tumor sites and lower invasiveness into normal healthy tissues. PTT relies on photothermal agents (PTAs), which generate heat by absorbing the near-infrared (NIR) light and destroying cancer cells. Several PTT agents remain longer in the reticuloendothelial system (RES) and induce toxicity, restricting their use in the biomedical field. To overcome this problem, the usage of biodegradable nano-photothermal agents is required. This review has discussed the PTT mechanism of action and different types of novel bio-nanomaterials used for PTT. We also focussed on the combinatorial effects of PTT with other cancer therapies and their effect on human health. The role of LED lights and mild hypothermia in PTT has been discussed briefly in this review.
Collapse
Affiliation(s)
| | | | | | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana-502285, India
| |
Collapse
|
9
|
Chen Y, Han Z, Zhang L, Gao C, Wei J, Yang X, Han Y, Li Y, Zhang C, Wei Y, Dong J, Xun W, Sun W, Zhang T, Zhang H, Chen J, Yuan P. TIMELESS promotes reprogramming of glucose metabolism in oral squamous cell carcinoma. J Transl Med 2024; 22:21. [PMID: 38178094 PMCID: PMC10768318 DOI: 10.1186/s12967-023-04791-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/09/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC), the predominant malignancy of the oral cavity, is characterized by high incidence and low survival rates. Emerging evidence suggests a link between circadian rhythm disruptions and cancer development. The circadian gene TIMELESS, known for its specific expression in various tumors, has not been extensively studied in the context of OSCC. This study aims to explore the influence of TIMELESS on OSCC, focusing on cell growth and metabolic alterations. METHODS We analyzed TIMELESS expression in OSCC using western blot, immunohistochemistry, qRT-PCR, and data from The Cancer Genome Atlas (TCGA) and the Cancer Cell Line Encyclopedia (CCLE). The role of TIMELESS in OSCC was examined through clone formation, MTS, cell cycle, and EdU assays, alongside subcutaneous tumor growth experiments in nude mice. We also assessed the metabolic impact of TIMELESS by measuring glucose uptake, lactate production, oxygen consumption, and medium pH, and investigated its effect on key metabolic proteins including silent information regulator 1 (SIRT1), hexokinase 2 (HK2), pyruvate kinase isozyme type M2 (PKM2), recombinant lactate dehydrogenase A (LDHA) and glucose transporter-1 (GLUT1). RESULTS Elevated TIMELESS expression in OSCC tissues and cell lines was observed, correlating with reduced patient survival. TIMELESS overexpression enhanced OSCC cell proliferation, increased glycolytic activity (glucose uptake and lactate production), and suppressed oxidative phosphorylation (evidenced by reduced oxygen consumption and altered pH levels). Conversely, TIMELESS knockdown inhibited these cellular and metabolic processes, an effect mirrored by manipulating SIRT1 levels. Additionally, SIRT1 was positively associated with TIMELESS expression. The expression of SIRT1, HK2, PKM2, LDHA and GLUT1 increased with the overexpression of TIMELESS levels and decreased with the knockdown of TIMELESS. CONCLUSION TIMELESS exacerbates OSCC progression by modulating cellular proliferation and metabolic pathways, specifically by enhancing glycolysis and reducing oxidative phosphorylation, largely mediated through the SIRT1 pathway. This highlights TIMELESS as a potential target for OSCC therapeutic strategies.
Collapse
Affiliation(s)
- Yafan Chen
- Department of Nuclear Medicine, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Zhengyang Han
- Department of Clinical Laboratory, Sinopharm Dongfeng General Hospital, Hubei University of Medicine, Shiyan, 442008, Hubei, China
| | - Le Zhang
- Department of Interventional Radiology and Pain Treatment, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, Shaanxi, China
| | - Caihong Gao
- Xi'an Physical Education University, Xi'an, 710068, Shaanxi, China
| | - Jingyi Wei
- The First Clinical Medical College, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Xuyuan Yang
- School of Nursing and Rehabilitation, Xi'an Medical University, Xi'an, 710021, Shaanxi, China
| | - Yabing Han
- Medical College of Ankang University, Ankang, 725000, Shaanxi, China
| | - Yunbo Li
- Department of Nuclear Medicine, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Chunmei Zhang
- Department of Nuclear Medicine, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Yixin Wei
- Department of Nuclear Medicine, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Jiaqi Dong
- The Second Clinical Medical School, Shaanxi University of Chinese Medicine, Xianyang, 712046, Shaanxi, China
| | - Wenxing Xun
- Department of Stomatology, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Weifu Sun
- Department of Stomatology, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Taotao Zhang
- Department of Stomatology, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Hui Zhang
- Department of Ultrasound Diagnosis, Xi'an Children's Hospital, 69 West Park Lane, Xi'an, 710002, Shaanxi, China.
| | - Jingtao Chen
- Department of Stomatology, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| | - Peng Yuan
- Department of Nuclear Medicine, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
10
|
Ning B, Mei Y. LAMA3 Promotes Tumorigenesis of Oral Squamous Cell Carcinoma by METTL3-Mediated N6-Methyladenosine Modification. Crit Rev Immunol 2024; 44:49-59. [PMID: 38305336 DOI: 10.1615/critrevimmunol.2023051066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Laminin subunit alpha 3 (LAMA3) is a cancer regulator. However, its effects and regulatory pathways in oral squamous cell carcinoma (OSCC) progression remain unknown. This research aimed to determine the influence of LAMA3 regulation via methyltransferase-like 3 (METTL3) on OSCC progression. Using quantitative real-time polymerase chain reaction and bioinformatics analysis, the expression levels of LAMA3 and METTL3 in OSCC tissues were examined. The functional roles of LAMA3 and METTL3 were analyzed using cell functional experiments. Using methylated RNA immunoprecipitation and mRNA stability assays, LAMA3 and METTL3 regulation was investigated. In OSCC tissues, LAMA3 was upregulated. LAMA3 inhibition hampered OSCC cell proliferation, invasion, and migration while its overexpression facilitated OSCC cell progression. METTL3 serves as a crucial upstream regulator of LAMA3 in OSCC and upregulates LAMA3 expression via an m6A-dependent mechanism. The low METTL3 expression partially restored the enhanced malignant phenotype induced by LAMA3 overexpression. Our findings indicate that METTL3 and LAMA3 act as pro-oncogenic factors in OSCC, with METTL3 promoting OSCC malignancy via m6A modification-dependent stabilization of LAMA3 transcripts, representing a novel regulatory mechanism in OSCC.
Collapse
Affiliation(s)
- Baoshan Ning
- Department of Stomatology, Wuhan Dongxihu District People's Hospital, Wuhan 430040, Hubei, China
| | - Yine Mei
- Department of Stomatology, Wuhan Dongxihu District People's Hospital, Wuhan 430040, Hubei, China
| |
Collapse
|
11
|
Li Y, Li R, Qin H, He H, Li S. OTUB1's role in promoting OSCC development by stabilizing RACK1 involves cell proliferation, migration, invasion, and tumor-associated macrophage M1 polarization. Cell Signal 2023; 110:110835. [PMID: 37532135 DOI: 10.1016/j.cellsig.2023.110835] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/19/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023]
Abstract
Ovarian tumor domain, ubiquitin aldehyde binding 1 (OTUB1), a deubiquitinating enzyme known to regulate the stability of downstream proteins, has been reported to regulate various cancers tumorigenesis, yet its direct effects on oral squamous cell carcinoma (OSCC) progression are unclear. Bioinformatics analysis was performed to screen for genes of interest, and in vitro and in vivo studies were carried out to investigate the function and mechanism of OTUB1 in OSCC. We found that OTUB1 was abnormally elevated in OSCC tissues and positively associated with the pathological stage and tumor stage. Knockdown of OTUB1 impaired the malignance of OSCC cells - suppressed cell proliferation, invasion, migration, and xenografted tumor growth. OTUB1 silencing also drove tumor-associated macrophage M1 polarization but suppressed M2 polarization, and the induction of M1 polarization inhibited the survival of OSCC cells. However, OTUB1 overexpression exerted the opposite effects. Furthermore, the protein network that interacted with the OTUB1 protein was constructed based on the GeneMANIA website. Receptor for activated C kinase 1 (RACK1), a facilitator of OSCC progression, was identified as a potential target of the OTUB1 protein. We revealed that OTUB1 positively regulated RACK1 expression and inhibited RACK1 ubiquitination. Additionally, RACK1 upregulation reversed the effects of OTUB1 knockdown on OSCC progression. Overall, we demonstrated that OTUB1 might regulate OSCC progression by maintaining the stability of the RACK1 protein. These findings highlight the potential roles of the OTUB1/RACK1 axis as a potential therapeutic target in OSCC.
Collapse
Affiliation(s)
- Yunyun Li
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Department of Stomatology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ruizhe Li
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Department of Pathology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hui Qin
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Department of Pathology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongliu He
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Department of Pathology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shanshan Li
- Department of Pathology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China; Department of Pathology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
12
|
Wen Z, Zhang Y, Wang X, Wu Y, Mao J, Li Q, Gong S. THBS1-Mediated Degradation of Collagen via the PI3K/AKT Pathway Facilitates the Metastasis and Poor Prognosis of OSCC. Int J Mol Sci 2023; 24:13312. [PMID: 37686118 PMCID: PMC10488045 DOI: 10.3390/ijms241713312] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/12/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a prevalent form of malignant tumor, characterized by a persistently high incidence and mortality rate. The extracellular matrix (ECM) plays a crucial role in the initiation, progression, and diverse biological behaviors of OSCC, facilitated by mechanisms such as providing structural support, promoting cell migration and invasion, regulating cell morphology, and modulating signal transduction. This study investigated the involvement of ECM-related genes, particularly THBS1, in the prognosis and cellular behavior of OSCC. The analysis of ECM-related gene data from OSCC samples identified 165 differentially expressed genes forming two clusters with distinct prognostic outcomes. Seventeen ECM-related genes showed a significant correlation with survival. Experimental methods were employed to demonstrate the impact of THBS1 on proliferation, migration, invasion, and ECM degradation in OSCC cells. A risk-prediction model utilizing four differentially prognostic genes demonstrated significant predictive value in overall survival. THBS1 exhibited enrichment of the PI3K/AKT pathway, indicating its potential role in modulating OSCC. In conclusion, this study observed and verified that ECM-related genes, particularly THBS1, have the potential to influence the prognosis, biological behavior, and immunotherapy of OSCC. These findings hold significant implications for enhancing survival outcomes and providing guidance for precise treatment of OSCC.
Collapse
Affiliation(s)
- Zhihao Wen
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
| | - Yuxiao Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiangyao Wang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yaxin Wu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Jing Mao
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Qilin Li
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Shiqiang Gong
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Z.W.); (Y.Z.); (X.W.); (Y.W.); (J.M.)
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| |
Collapse
|
13
|
Recent Clinical and Preclinical Advances in External Stimuli-Responsive Therapies for Head and Neck Squamous Cell Carcinoma. J Clin Med 2022; 12:jcm12010173. [PMID: 36614974 PMCID: PMC9821160 DOI: 10.3390/jcm12010173] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/14/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) has long been one of the most prevalent cancers worldwide; even though treatments such as surgery, chemotherapy, radiotherapy and immunotherapy have been proven to benefit the patients and prolong their survival time, the overall five-year survival rate is still below 50%. Hence, the development of new therapies for better patient management is an urgent need. External stimuli-responsive therapies are emerging therapies with promising antitumor effects; therapies such as photodynamic (PDT) and photothermal therapies (PTT) have been tested clinically in late-stage HNSCC patients and have achieved promising outcomes, while the clinical translation of sonodynamic therapy (SDT), radiodynamic therapy (RDT), microwave dynamic/thermodynamic therapy, and magnetothermal/magnetodynamic therapy (MDT/MTT) still lag behind. In terms of preclinical studies, PDT and PTT are also the most extensively studied therapies. The designing of nanoparticles and combinatorial therapies of PDT and PTT can be referenced in designing other stimuli-responsive therapies in order to achieve better antitumor effects as well as less toxicity. In this review, we consolidate the advancements and limitations of various external stimuli-responsive therapies, as well as critically discuss the prospects of this type of therapies in HNSCC treatments.
Collapse
|
14
|
Li Y, Su Y, Li Z, Chen Y. Supramolecular Combination Cancer Therapy Based on Macrocyclic Supramolecular Materials. Polymers (Basel) 2022; 14:polym14224855. [PMID: 36432982 PMCID: PMC9696801 DOI: 10.3390/polym14224855] [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: 10/06/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/15/2022] Open
Abstract
Supramolecular combination therapy adopts supramolecular materials to design intelligent drug delivery systems with different strategies for cancer treatments. Thereinto, macrocyclic supramolecular materials play a crucial role in encapsulating anticancer drugs to improve anticancer efficiency and decrease toxicity towards normal tissue by host-guest interaction. In general, chemotherapy is still common therapy for solid tumors in clinics. However, supramolecular combination therapy can overcome the limitations of the traditional single-drug chemotherapy in the laboratory findings. In this review, we summarized the combination chemotherapy, photothermal chemotherapy, and gene chemotherapy based on macrocyclic supramolecular materials. Finally, the application prospects in supramolecular combination therapy are discussed.
Collapse
|