1
|
Khutale G, Andhari S, Gupta R, Aland G, Banerjee S, Todkar K, Pore M, Khobragade V, D'Souza A, Kale N, Vasudevan A, Bharde A, Jayant S, Patil Y, Khandare J. A multicomponent nanosystem for capturing circulating tumor cells from cancer patients with PD-L1 as an immunotherapy oncotarget. J Mater Chem B 2024; 12:10973-10982. [PMID: 39348156 DOI: 10.1039/d4tb01360c] [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: 10/01/2024]
Abstract
Capturing circulating tumor cells (CTCs) from the peripheral blood of cancer patients, where they are disseminated among billions of other blood cells, is one of the most daunting challenge. We report OncoDiscover®, a multicomponent nano-system consisting of iron oxide (Fe3O4) nanoparticles (NPs), polyamidoamine generation 4 dendrimers (PAMAM-G4-NH2), graphene oxide (GO) sheets and an anti-epithelial cell adhesion molecule (anti-EpCAM) antibody (Fe-GSH-PAMAM-GO-EpCAM) for the selective and precise capture of CTCs. We further evaluated this system for therapeutically important oncotargets, exemplifying overexpression of the programmed death ligand 1 (PD-L1) as a functional assay on CTCs in cancer patients. We retrospectively evaluated 134 cancer patients for the presence of CTCs and 113 (84%) showed the presence of CTCs. About 62 (55%) of the CTC +ve patients showed CTCs with PD-L1 expression. The personalized cancer detection demonstrated by the OncoDiscover® CTC analysis tool is highly relevant for cancer monitoring and treatment outcomes.
Collapse
Affiliation(s)
- Ganesh Khutale
- Actorius Innovations and Research, Pune 411057, India.
- OneCell Diagnostics, Pune 411057, India
| | - Saloni Andhari
- Actorius Innovations and Research, Pune 411057, India.
- OneCell Diagnostics, Pune 411057, India
| | - Rituja Gupta
- School of Health Sciences and Technology, Dr Vishwanath Karad MIT World Peace University, Kothrud, Pune 411038, India
| | | | | | - Kiran Todkar
- Actorius Innovations and Research, Pune 411057, India.
| | - Milind Pore
- Actorius Innovations and Research, Pune 411057, India.
| | | | - Alain D'Souza
- Actorius Innovations and Research, Pune 411057, India.
- OneCell Diagnostics, Pune 411057, India
| | - Narendra Kale
- Center for Nanomedicine, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | | | | | - Sreeja Jayant
- Actorius Innovations and Research, Pune 411057, India.
| | - Yuvraj Patil
- School of Health Sciences and Technology, Dr Vishwanath Karad MIT World Peace University, Kothrud, Pune 411038, India
| | - Jayant Khandare
- Actorius Innovations and Research, Pune 411057, India.
- OneCell Diagnostics, Pune 411057, India
- School of Health Sciences and Technology, Dr Vishwanath Karad MIT World Peace University, Kothrud, Pune 411038, India
- Actorius Innovations and Research, Simi Valley, CA 93063, USA
- School of Consciousness, Dr Vishwanath Karad MIT World Peace University, Kothrud, Pune 411038, India
| |
Collapse
|
2
|
Mesquita F, de Oliveira FL, da Silva EL, Brito DM, de Moraes ME, Souza PF, Montenegro RC. Synthetic Peptides Induce Human Colorectal Cancer Cell Death via Proapoptotic Pathways. ACS OMEGA 2024; 9:43252-43263. [PMID: 39464451 PMCID: PMC11500374 DOI: 10.1021/acsomega.4c08194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/29/2024]
Abstract
Cancer resistance to drugs and chemotherapy is a problem faced by public health systems worldwide. Repositioning antimicrobial peptides could be an efficient strategy to overcome that problem. This study aimed at repurposing antimicrobial peptides PepGAT and PepKAA for cancer treatment. After screening against several cancers, PepGAT and PepKAA presented IC50 values of 125.42 and 40.51 μM at 72 h toward colorectal cancer (CRC) cells. The mechanisms of action revealed that both peptides induced cell cycle arrest in G2/M and drove HCT-116 cells to death by triggering apoptosis. qPCR analysis revealed that peptides modulated gene expression in apoptosis, corroborating the data from caspase 3/7 and flow cytometry experiments. Yet, peptides induced ROS overaccumulation and increased membrane permeabilization, pore formation, and loss of internal content, leading to death. Additionally, peptides were able to inhibit cell invasion. Previous studies from the same group attested to no toxicity to normal human cells. Thus, PepGAT and PepKAA have great potential as anticancer molecules.
Collapse
Affiliation(s)
- Felipe
P. Mesquita
- Pharmacogenetics
Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Francisco L. de Oliveira
- Pharmacogenetics
Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Emerson L. da Silva
- Pharmacogenetics
Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Daiane M.S. Brito
- Pharmacogenetics
Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Maria E.A. de Moraes
- Pharmacogenetics
Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
| | - Pedro F.N. Souza
- Pharmacogenetics
Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
- Cearense
Foundation to Support Scientific and Technological Development, Fortaleza 60822-131, Brazil
| | - Raquel C. Montenegro
- Pharmacogenetics
Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE 60430-275, Brazil
- Red
Latinoamericana de Implementación y Validación de guias
clinicas Farmacogenomicas (RELIVAF), Madrid 28015, Spain
| |
Collapse
|
3
|
Pan S, Yuan H, Zhai Q, Zhang Y, He H, Yin T, Tang X, Gou J. The journey of nanoparticles in the abdominal cavity: Exploring their in vivo fate and impact factors. J Control Release 2024; 376:266-285. [PMID: 39396710 DOI: 10.1016/j.jconrel.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/15/2024]
Abstract
Peritoneal carcinomatosis (PC) is caused by metastasis of primary tumor cells from intra-abdominal organs to the peritoneal surface. Intraperitoneal (IP) chemotherapy allows close contact of high concentrations of therapeutic agents with cancer cells in the peritoneal cavity to prolong patient survival. However, conventional IP chemotherapy is prone to rapid elimination from the peritoneal cavity and lacks specificity towards cancer cells. To address these challenges, there is an imperative demand for exploiting novel drug delivery systems to enhance drug retention in the peritoneal cavity and target PC cells. Therefore, in this review, we first recapitulate the physiological structures and barriers associated with IP drug delivery, highlighting the in vivo fate of nanoparticles (NPs) after IP administration. Furthermore, the influence of physicochemical properties (particle size, charge, surface modification, and carrier composition) on the in vivo fate of NPs is discussed. Perspectives on the rational design of NPs for IP therapy and recent clinical progress are also provided.
Collapse
Affiliation(s)
- Shu Pan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Haoyang Yuan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Qiyao Zhai
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China.
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China.
| |
Collapse
|
4
|
Kitagawa K, Mitoro A, Suzuki H, Tomooka F, Asada S, Hanatani JI, Motokawa Y, Iwata T, Osaki Y, Takeda M, Yoshiji H. Role of Liquid-Based Cytology in the Endoscopic Diagnosis of Pancreatic Ductal Adenocarcinoma. J Clin Med 2024; 13:6148. [PMID: 39458098 PMCID: PMC11509073 DOI: 10.3390/jcm13206148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/05/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Recently, endoscopic ultrasound-guided tissue acquisition (EUS-TA) has been widely used to diagnose pancreatic ductal adenocarcinoma (PDAC). The histological examination of core tissues acquired using novel biopsy needles is the primary diagnostic approach for patients with PDAC. However, in patients with early-stage PDAC, such as Stages 0 and I, EUS-TA can be challenging, and its diagnostic accuracy may be limited. This presents a clinical dilemma: The earlier that clinicians attempt to accurately diagnose PDAC, the more difficult it becomes to do so using EUS-TA. Liquid-based cytology (LBC) is a technique for preparing pathological specimens from liquefied cytology specimens by placing the collected material in a special fixative preservative fluid. LBC offers advantages, such as specimen optimization with reduced blood interference, a high cell-collection rate, and the simplicity of the procedure in the endoscopy room. The use of LBC may improve diagnostic accuracy, particularly for early-stage PDAC. Therefore, we emphasize that cytology remains a valuable tool for the endoscopic diagnosis of PDAC. In this review, we discuss the role of LBC in the endoscopic diagnosis of PDAC.
Collapse
Affiliation(s)
- Koh Kitagawa
- Department of Gastroenterology, Nara Medical University, Nara 634-8522, Japan
| | - Akira Mitoro
- Division of Endoscopy, Nara Medical University, Nara 634-8522, Japan
| | - Hisae Suzuki
- Department of Diagnostic Pathology, Nara Medical University, Nara 634-8522, Japan
| | - Fumimasa Tomooka
- Department of Gastroenterology, Nara Medical University, Nara 634-8522, Japan
| | - Shohei Asada
- Department of Gastroenterology, Nara Medical University, Nara 634-8522, Japan
| | - Jun-Ichi Hanatani
- Department of Gastroenterology, Nara Medical University, Nara 634-8522, Japan
| | - Yuki Motokawa
- Department of Gastroenterology, Nara Medical University, Nara 634-8522, Japan
| | - Tomihiro Iwata
- Department of Gastroenterology, Nara Medical University, Nara 634-8522, Japan
| | - Yui Osaki
- Department of Gastroenterology, Nara Medical University, Nara 634-8522, Japan
| | - Maiko Takeda
- Department of Diagnostic Pathology, Nara Medical University, Nara 634-8522, Japan
| | - Hitoshi Yoshiji
- Department of Gastroenterology, Nara Medical University, Nara 634-8522, Japan
| |
Collapse
|
5
|
Wen Z, Qin S, Huang H, Xia X, Zhang W, Wu W. Functional exosomes modified with chitosan effectively alleviate anthracycline-induced cardiotoxicity. Int J Biol Macromol 2024; 277:134495. [PMID: 39111472 DOI: 10.1016/j.ijbiomac.2024.134495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/14/2024] [Accepted: 08/02/2024] [Indexed: 08/10/2024]
Abstract
Anthracyclines belong to a class of anti-tumor antibiotics, and their severe cardiotoxicity significantly limits their clinical use. Exosomes play key roles in intercellular communication, characterized by high biocompatibility and specific tissue and organ homing effects. In this study, doxorubicin, an anthracycline anticancer drug widely used in clinical chemotherapy, was selected as a model drug. To address the significant cardiotoxicity associated with doxorubicin, tumor exosomes are utilized as drug carriers. The homing effect of autologous exosomes enhances drug uptake by tumor cells and reduces cardiotoxicity. To enhance the stability of exosomes, improve therapeutic effectiveness, and reduce toxic side effects, chitosan was utilized to modify the surface of exosomes. Chitosan has a specific anti-tumor effect because it can target the CD44 receptor of tumor stem cells and interact with tumor cells through charge adsorption. Through in vitro cell experiments, in vivo pharmacokinetic experiments, and an in situ ectopic nude mouse tumor model, the study demonstrated that chitosan-modified tumor exosomes significantly alleviated the severe cardiotoxicity of doxorubicin, while also showing remarkable anti-tumor efficacy. This study introduces a novel approach to reduce the adverse side effects of anthracycline chemotherapeutic drugs and presents a highly promising nanocarrier delivery system.
Collapse
Affiliation(s)
- Zhiwei Wen
- School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Shuiling Qin
- School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Huajie Huang
- School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Xingle Xia
- School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Wei Zhang
- School of Pharmacy, Guilin Medical University, Guilin 541199, China
| | - Wei Wu
- School of Pharmacy, Guilin Medical University, Guilin 541199, China; Guangxi Key Laboratory of Drug Discovery and Optimization, School of Pharmacy, Guilin Medical University, Guilin 541199, China.
| |
Collapse
|
6
|
Liu G, Zhu J, Zhang P, Zhang T, Cui Z, Jiao F, Le W, Li X, Chen B. Exploring the diagnostic and prognostic significance of circulating tumor cells in stage II-IV colorectal cancer using a nano-based detection method. J Chin Med Assoc 2024; 87:945-952. [PMID: 39164812 DOI: 10.1097/jcma.0000000000001143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a leading cause of cancer mortality globally, underscoring the urgency for a noninvasive and effective biomarker to enhance patient prognosis. Circulating tumor cells (CTCs), a potential marker for real-time tumor monitoring, are limited in clinical utility due to the low sensitivity of existing detection methods. Previously, we introduced a novel nano-based CTCs detection method that relies on the electrical properties of cell surfaces, thus eliminating the need for specific molecular biomarkers. In this study, we used this technique to evaluate the diagnostic and prognostic value of CTCs in stage II-IV CRC. METHODS A total of 194 participants were included, consisting of 136 CRC patients and 58 healthy individuals. The peripheral blood of the participants was collected, and CTC enumeration was performed utilizing the nano-based detection method that we newly developed. The receiver operating characteristic (ROC) curve and multivariate Cox proportional-hazards analysis were used to assess the effectiveness of CTCs for diagnosing CRC and predicting patient prognosis. RESULTS The nano-based method demonstrated an ability to differentiate CRC patients from healthy individuals with a sensitivity of 84.6% and a specificity of 94.8%. Furthermore, baseline CTC levels were predictive of progression-free survival (PFS) in CRC patients, with lower levels associated with longer PFS compared to higher levels (4.5 vs 8.0 months at 15 CTCs/mL, p = 0.016; 4.4 vs 8.0 months at 20 CTCs/mL, p = 0.028). We also explored the dynamic changes in the number of CTCs after 1 to 5 cycles of chemotherapy. Patients with increasing CTC levels typically experienced disease progression (PD), while those with decreasing levels often achieved a partial response (PR) or maintained stable disease (SD). These findings suggest that the dynamic fluctuations in CTC counts are closely tied to the clinical course of the disease. CONCLUSION Our study indicates the potential of nano-based CTCs detection in diagnosing and predicting outcomes for patients with stage II-IV CRC.
Collapse
Affiliation(s)
- Gang Liu
- Department of Throatic Surgery, East Hospital of Tongji University School of Medicine, Shanghai, China
| | - Jinfeng Zhu
- Department of Oncology, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Pengbo Zhang
- Zhihui Medical Technology (Shanghai) Co., Ltd., Shanghai, China
| | - Tingting Zhang
- Zhihui Medical Technology (Shanghai) Co., Ltd., Shanghai, China
| | - Zheng Cui
- Zhihui Medical Technology (Shanghai) Co., Ltd., Shanghai, China
| | - Fanglei Jiao
- Department of Throatic Surgery, East Hospital of Tongji University School of Medicine, Shanghai, China
| | - Wenjun Le
- Institute for Regenerative Medicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China
| | - Xiaofeng Li
- Department of Oncology, Affiliated Quanzhou First Hospital of Fujian Medical University, Quanzhou, China
| | - Bingdi Chen
- Institute for Regenerative Medicine, Shanghai East Hospital, The Institute for Biomedical Engineering & Nano Science, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
7
|
Cai Q, He Y, Zhou Y, Zheng J, Deng J. Nanomaterial-Based Strategies for Preventing Tumor Metastasis by Interrupting the Metastatic Biological Processes. Adv Healthc Mater 2024; 13:e2303543. [PMID: 38411537 DOI: 10.1002/adhm.202303543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/01/2024] [Indexed: 02/28/2024]
Abstract
Tumor metastasis is the primary cause of cancer-related deaths. The prevention of tumor metastasis has garnered notable interest and interrupting metastatic biological processes is considered a potential strategy for preventing tumor metastasis. The tumor microenvironment (TME), circulating tumor cells (CTCs), and premetastatic niche (PMN) play crucial roles in metastatic biological processes. These processes can be interrupted using nanomaterials due to their excellent physicochemical properties. However, most studies have focused on only one aspect of tumor metastasis. Here, the hypothesis that nanomaterials can be used to target metastatic biological processes and explore strategies to prevent tumor metastasis is highlighted. First, the metastatic biological processes and strategies involving nanomaterials acting on the TME, CTCs, and PMN to prevent tumor metastasis are briefly summarized. Further, the current challenges and prospects of nanomaterials in preventing tumor metastasis by interrupting metastatic biological processes are discussed. Nanomaterial-and multifunctional nanomaterial-based strategies for preventing tumor metastasis are advantageous for the long-term fight against tumor metastasis and their continued exploration will facilitate rapid progress in the prevention, diagnosis, and treatment of tumor metastasis. Novel perspectives are outlined for developing more effective strategies to prevent tumor metastasis, thereby improving the outcomes of patients with cancer.
Collapse
Affiliation(s)
- Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Yijia He
- School of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yang Zhou
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400037, China
| | - Jun Deng
- Institute of Burn Research, Southwest Hospital, State Key Lab of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| |
Collapse
|
8
|
Ai J, Huang Y, Yin Z, Deng Y, Yan L, Liao J, Liang G, Chen C, Chang Y, Xiao C, Zhou J, Zhu Z, Liu C, Jiang Z, Ning C, Wang Z. Sea Anemone-Inspired Conducting Polymer Sensing Platform for Integrated Detection of Tumor Protein Marker and Circulating Tumor Cell. Adv Healthc Mater 2024:e2401305. [PMID: 38767216 DOI: 10.1002/adhm.202401305] [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: 04/09/2024] [Indexed: 05/22/2024]
Abstract
Combining the detection of tumor protein markers with the capture of circulating tumor cells (CTCs) represents an ultra-promising approach for early tumor detection. However, current methodologies have not yet achieved the necessary low detection limits and efficient capture. Here, a novel polypyrrole nanotentacles sensing platform featuring anemone-like structures capable of simultaneously detecting protein biomarkers and capturing CTCs is introduced. The incorporation of nanotentacles significantly enhances the electrode surface area, providing abundant active sites for antibody binding. This enhancement allows detecting nucleus matrix protein22 and bladder tumor antigen with 2.39 and 3.12 pg mL-1 detection limit, respectively. Furthermore, the developed sensing platform effectively captures MCF-7 cells in blood samples with a detection limit of fewer than 10 cells mL-1, attributed to the synergistic multivalent binding facilitated by the specific recognition antibodies and the positive charge on the nanotentacles surface. This sensing platform demonstrates excellent detection capabilities and outstanding capture efficiency, offering a simple, accurate, and efficient strategy for early tumor detection.
Collapse
Affiliation(s)
- Jialuo Ai
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Yixuan Huang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Zhaoyi Yin
- School of Materials Science and Technology, Kunming University of Science and Technology, Kunming, 650093, China
| | - Yingshan Deng
- School of Life Sciences, South China Normal University, Guangzhou, 510631, P. R. China
| | - Ling Yan
- Department of Clinical Laboratory, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Jingwen Liao
- Interdisciplinary Plasma Engineering Centre, Guangzhou Institute of Advanced Technology, Guangzhou, 511458, P. R. China
| | - Guoyan Liang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangzhou, 510080, P. R. China
| | - Chong Chen
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangzhou, 510080, P. R. China
| | - Yunbing Chang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangzhou, 510080, P. R. China
| | - Cairong Xiao
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Jiale Zhou
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Zurong Zhu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Chengli Liu
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Zhuo Jiang
- College of Food Science, South China Agricultural University, Guangzhou, 510642, P. R. China
| | - Chengyun Ning
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- GuangDong Engineering Technology Research Center of Metallic Materials Surface Functionalization, South China University of Technology, Guangzhou, 510006, P. R. China
| | - Zhengao Wang
- School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510006, P. R. China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, P. R. China
- Research Center of Biomass 3D Printing Materials, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, P. R. China
| |
Collapse
|
9
|
Zenze M, Singh M. Receptor Targeting Using Copolymer-Modified Gold Nanoparticles for pCMV-Luc Gene Delivery to Liver Cancer Cells In Vitro. Int J Mol Sci 2024; 25:5016. [PMID: 38732235 PMCID: PMC11084699 DOI: 10.3390/ijms25095016] [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: 04/15/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/13/2024] Open
Abstract
The formulation of novel delivery protocols for the targeted delivery of genes into hepatocytes by receptor mediation is important for the treatment of liver-specific disorders, including cancer. Non-viral delivery methods have been extensively studied for gene therapy. Gold nanoparticles (AuNPs) have gained attention in nanomedicine due to their biocompatibility. In this study, AuNPs were synthesized and coated with polymers: chitosan (CS), and polyethylene glycol (PEG). The targeting moiety, lactobionic acid (LA), was added for hepatocyte-specific delivery. Physicochemical characterization revealed that all nano-formulations were spherical and monodispersed, with hydrodynamic sizes between 70 and 250 nm. Nanocomplexes with pCMV-Luc DNA (pDNA) confirmed that the NPs could bind, compact, and protect the pDNA from nuclease degradation. Cytotoxicity studies revealed that the AuNPs were well tolerated (cell viabilities > 70%) in human hepatocellular carcinoma (HepG2), embryonic kidney (HEK293), and colorectal adenocarcinoma (Caco-2) cells, with enhanced transgene activity in all cells. The inclusion of LA in the NP formulation was notable in the HepG2 cells, which overexpress the asialoglycoprotein receptor on their cell surface. A five-fold increase in luciferase gene expression was evident for the LA-targeted AuNPs compared to the non-targeted AuNPs. These AuNPs have shown potential as safe and suitable targeted delivery vehicles for liver-directed gene therapy.
Collapse
Affiliation(s)
| | - Moganavelli Singh
- Nano-Gene and Drug Delivery Laboratory, Discipline of Biochemistry, University of KwaZulu-Natal, Private Bag X54001, Durban 4000, South Africa;
| |
Collapse
|
10
|
Furukawa N, Yang W, Chao AR, Patil A, Mirando AC, Pandey NB, Popel AS. Chemokine-derived oncolytic peptide induces immunogenic cancer cell death and significantly suppresses tumor growth. Cell Death Discov 2024; 10:161. [PMID: 38565596 PMCID: PMC10987543 DOI: 10.1038/s41420-024-01932-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/04/2024] Open
Abstract
Chemokinostatin-1 (CKS1) is a 24-mer peptide originally discovered as an anti-angiogenic peptide derived from the CXCL1 chemokine. Here, we demonstrate that CKS1 acts not only as an anti-angiogenic peptide but also as an oncolytic peptide due to its structural and physical properties. CKS1 induced both necrotic and apoptotic cell death specifically in cancer cells while showing minimal toxicity in non-cancerous cells. Mechanistically, CKS1 disrupted the cell membrane of cancer cells quickly after treatment and activated the apoptotic pathway at later time points. Furthermore, immunogenic molecules were released from CKS1-treated cells, indicating that CKS1 induces immunogenic cell death. CKS1 effectively suppressed tumor growth in vivo. Collectively, these data demonstrate that CKS1 functions as an oncolytic peptide and has a therapeutic potential to treat cancer.
Collapse
Affiliation(s)
- Natsuki Furukawa
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Wendy Yang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alex R Chao
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Akash Patil
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Adam C Mirando
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Niranjan B Pandey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
11
|
Bahremand K, Aghaz F, Bahrami K. Enhancing Cisplatin Efficacy with Low Toxicity in Solid Breast Cancer Cells Using pH-Charge-Reversal Sericin-Based Nanocarriers: Development, Characterization, and In Vitro Biological Assessment. ACS OMEGA 2024; 9:14017-14032. [PMID: 38560009 PMCID: PMC10976391 DOI: 10.1021/acsomega.3c09361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 01/23/2024] [Accepted: 03/04/2024] [Indexed: 04/04/2024]
Abstract
Platinum-based chemotherapeutic agents are widely employed in cancer treatment because of their effectiveness in targeting DNA. However, this indiscriminate action often affects both cancerous and normal cells, leading to severe side effects and highlighting the need for innovative approaches in achieving precise drug delivery. Nanotechnology presents a promising avenue for addressing these challenges. Protein-based nanocarriers exhibit promising capabilities in the realm of cancer drug delivery with silk sericin nanoparticles standing out as a leading contender. This investigation focuses on creating a sericin-based nanocarrier (SNC) featuring surface charge reversal designed to effectively transport cisplatin (Cispt-SNC) into MCF-7 breast cancer cells. Utilizing AutoDock4.2, our molecular docking analyses identified key amino acids and revealed distinctive conformational clusters, providing insights into the drug-protein interaction landscape and highlighting the potential of sericin as a carrier for controlled drug release. The careful optimization and fabrication of sericin as the carrier material were achieved through flash nanoprecipitation, a straightforward and reproducible method that is devoid of intricate equipment. The physicochemical properties of SNCs and Cispt-SNCs, particularly concerning size, surface charge, and morphology, were evaluated using dynamic light scattering (DLS) and scanning electron microscopy (SEM). Chemical and conformational analyses of the nanocarriers were conducted using Fourier-transform infrared spectroscopy (FTIR) and circular dichroism (CD), and elemental composition analysis was performed through energy-dispersive X-ray spectroscopy (EDX). This approach aimed to achieve the smallest nanoparticle size for Cispt-SNCs (180 nm) and high drug encapsulation efficiency (84%) at an optimal sericin concentration of 0.1% (w/v), maintaining a negative net charge at a physiological pH (7.4). Cellular uptake and cytotoxicity were investigated in MCF-7 breast cancer cells. SNCs demonstrated stability and exhibited a pH-dependent drug release behavior, aligning with the mildly acidic tumor microenvironment (pH 6.0-7.0). Efficient cellular uptake of Cispt-SNC, along with DNA fragmentation and chromatin condensation, was found at pH 6, leading to cell apoptosis. These results collectively indicate the potential of SNCs for achieving controlled drug release in a tumor-specific context. Our in vitro studies reveal the cytotoxicity of both cisplatin and Cispt-SNCs on MCF-7 cells. Cisplatin significantly reduced cell viability at 10 μM concentration (IC50), and the unique combination of sericin and cisplatin showcased enhanced cell viability compared to cisplatin alone, suggesting that controlled drug release is indicated by a gradient decrease in cell viability and highlighting SNCs as promising carriers. The study underscores the promise of protein-based nanocarriers in advancing targeted drug delivery for cancer therapy.
Collapse
Affiliation(s)
- Kiana Bahremand
- Nano Drug Delivery
Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Faranak Aghaz
- Nano Drug Delivery
Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Kiumars Bahrami
- Nanoscience and Nanotechnology
Research Center (NNRC), Razi University, Kermanshah 67144-14971, Iran
| |
Collapse
|
12
|
Kawasaki H, Hariyama T, Kosugi I, Meguro S, Iwata F, Shimizu K, Magata Y, Iwashita T. Human induced pluripotent stem cells are resistant to human cytomegalovirus infection primarily at the attachment level due to the reduced expression of cell-surface heparan sulfate. J Virol 2024; 98:e0127823. [PMID: 38345384 PMCID: PMC10949504 DOI: 10.1128/jvi.01278-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 01/16/2024] [Indexed: 03/20/2024] Open
Abstract
Cytomegalovirus (CMV), a type of herpes virus, is the predominant cause of congenital anomalies due to intrauterine infections in humans. Adverse outcomes related to intrauterine infections with human cytomegalovirus (HCMV) vary widely, depending on factors such as fetal infection timing, infection route, and viral virulence. The precise mechanism underlying HCMV susceptibility remains unclear. In this study, we compared the susceptibility of neonatal human dermal fibroblast cells (NHDFCs) and human induced pluripotent stem cells (hiPSCs) derived from NHDFCs, which are genetically identical to HCMV, using immunostaining, microarray, in situ hybridization, quantitative PCR, and scanning electron microscopy. These cells were previously used to compare CMV susceptibility, but the underlying mechanisms were not fully elucidated. HCMV susceptibility of hiPSCs was significantly lower in the earliest phase. No shared gene ontologies were observed immediately post-infection between the two cell types using microarray analysis. Early-stage expression of HCMV antigens and the HCMV genome was minimal in immunostaining and in in situ hybridization in hiPSCs. This strongly suggests that HCMV does not readily bind to hiPSC surfaces. Scanning electron microscopy performed using the NanoSuit method confirmed the scarcity of HCMV particles on hiPSC surfaces. The zeta potential and charge mapping of the charged surface in NHDFCs and hiPSCs exhibited minimal differences when assessed using zeta potential analyzer and scanning ion conductance microscopy; however, the expression of heparan sulfate (HS) was significantly lower in hiPSCs compared with that in NHDFCs. Thus, HS expression could be a primary determinant of HCMV resistance in hiPSCs at the attachment level. IMPORTANCE Numerous factors such as attachment, virus particle entry, transcription, and virus particle egress can affect viral susceptibility. Since 1984, pluripotent cells are known to be CMV resistant; however, the exact mechanism underlying this resistance remains elusive. Some researchers suggest inhibition in the initial phase of HCMV binding, while others have suggested the possibility of a sufficient amount of HCMV entering the cells to establish latency. This study demonstrates that HCMV particles rarely attach to the surfaces of hiPSCs. This is not due to limitations in the electrostatic interactions between the surface of hiPSCs and HCMV particles, but due to HS expression. Therefore, HS expression should be recognized as a key factor in determining the susceptibility of HCMV in congenital infection in vitro and in vivo. In the future, drugs targeting HS may become crucial for the treatment of congenital CMV infections. Thus, further research in this area is warranted.
Collapse
Affiliation(s)
- Hideya Kawasaki
- Institute for NanoSuit Research, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Takahiko Hariyama
- Institute for NanoSuit Research, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Isao Kosugi
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Shiori Meguro
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Futoshi Iwata
- Research Institute of Electronics, Shizuoka University, Hamamatsu, Shizuoka, Japan
| | - Kosuke Shimizu
- Department of Molecular Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Yasuhiro Magata
- Department of Molecular Imaging, Preeminent Medical Photonics Education and Research Center, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Toshihide Iwashita
- Department of Regenerative and Infectious Pathology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| |
Collapse
|
13
|
El-Ghannam G, Moawad M, Abo-Elfadl MT, Elfeky SA. Beetroot extract@chitosan nanocomposite as a promising approach towards cancer therapy. Int J Biol Macromol 2024; 261:129700. [PMID: 38278395 DOI: 10.1016/j.ijbiomac.2024.129700] [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: 09/17/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 01/28/2024]
Abstract
The exceptional antioxidant properties of beetroot (BR) and the cancer antiproliferative effects of chitosan nanoparticles (CS NP) have led to the synthesis of a BR@CS nanocomposite (NC) in this study. The novel BR@CS NC was applied to human epithelial colorectal adenocarcinoma (Caco-2), human epithelial ductal breast carcinoma (T-47D), and human epithelial lung carcinoma (A549) cells. SEM characterization of CS NP revealed a variety of particle shapes ranging from 20 to 58 nm in diameter. UV-VIS analysis confirmed the formation of the BR@CS NC, while FTIR analysis demonstrated strong hydrogen bonds between CS NP and BR. These bonds reduced the positive surface charge of CS NP, as indicated by zeta potential analysis. When applied to cancer cell lines at a concentration of 250 μg/mL, the BR@CS NC successfully eradicated 89 % of A549, 88 % of T-47D, and 83 % of Caco-2 cell lines. The cell death mode exhibited extensive, apoptotic, and massive necrotic changes in all cell lines treated with BR@CS NC. Caspase 3 (CasP3) and P53 levels were elevated in BR@CS NC-treated cells. This study merges BR's antioxidant and anti-inflammatory properties with the antiangiogenic mechanism and inhibition of tumors by CS NP, resulting in a unique and innovative strategy for cancer treatment.
Collapse
Affiliation(s)
- Gamal El-Ghannam
- National Institute of Laser Enhanced Sciences (NILES), Department of Laser Applications in Metrology, Photochemistry, and Agriculture (LAMPA), Cairo University, 12613 Giza, Egypt.
| | - Mahmoud Moawad
- Department of Surgical Pathology, National Cancer Institute, Cairo University, Egypt
| | - Mahmoud T Abo-Elfadl
- Biochemistry Department, Biotechnology Research Institute, National Research Centre, Cairo 12622, Egypt; Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Cairo 12622, Egypt
| | - Souad A Elfeky
- National Institute of Laser Enhanced Sciences (NILES), Department of Laser Applications in Metrology, Photochemistry, and Agriculture (LAMPA), Cairo University, 12613 Giza, Egypt.
| |
Collapse
|
14
|
Amer L, Retout M, Jokerst JV. Activatable prodrug for controlled release of an antimicrobial peptide via the proteases overexpressed in Candida albicans and Porphyromonas gingivalis. Theranostics 2024; 14:1781-1793. [PMID: 38389835 PMCID: PMC10879876 DOI: 10.7150/thno.91165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 02/11/2024] [Indexed: 02/24/2024] Open
Abstract
Candida albicans and Porphyromonas gingivalis are prevalent in the subgingival area where the frequency of fungal colonization increases with periodontal disease. Candida's transition to a pathogenic state and its interaction with P. gingivalis exacerbate periodontal disease severity. However, current treatments for these infections differ, and combined therapy remains unexplored. This work is based on an antimicrobial peptide that is therapeutic and induces a color change in a nanoparticle reporter. Methods: We built and characterized two enzyme-activatable prodrugs to treat and detect C. albicans and P. gingivalis via the controlled release of the antimicrobial peptide. The zwitterionic prodrug quenches the antimicrobial peptide's activity until activation by a protease inherent to the pathogens (SAP9 for C. albicans and RgpB for P. gingivalis). The toxicity of the intact prodrugs was evaluated against fungal, bacterial, and mammalian cells. Therapeutic efficacy was assessed through microscopy, disk diffusion, and viability assays, comparing the prodrug to the antimicrobial peptide alone. Finally, we developed a colorimetric detection system based on the aggregation of plasmonic nanoparticles. Results: The intact prodrugs showed negligible toxicity to cells absent a protease trigger. The therapeutic impact of the prodrugs was comparable to that of the antimicrobial peptide alone, with a minimum inhibitory concentration of 3.1 - 16 µg/mL. The enzymatic detection system returned a detection limit of 10 nM with gold nanoparticles and 3 nM with silver nanoparticles. Conclusion: This approach offers a convenient and selective protease sensing and protease-induced treatment mechanism based on bioinspired antimicrobial peptides.
Collapse
Affiliation(s)
- Lubna Amer
- Program in Materials Science and Engineering, University of California, San Diego, La Jolla, CA 92093, United States
| | - Maurice Retout
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, United States
| | - Jesse V. Jokerst
- Program in Materials Science and Engineering, University of California, San Diego, La Jolla, CA 92093, United States
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, United States
- Department of Radiology, University of California, San Diego, La Jolla, CA 92093, United States
| |
Collapse
|
15
|
Xue K, Zhao Y, Sun S, Li Y, Qi Z. A near-infrared aggregation-induced emission photosensitizer targeting mitochondria for depleting Cu 2+ to trigger light-activated cancer cells oncosis. Bioorg Chem 2024; 143:107020. [PMID: 38176374 DOI: 10.1016/j.bioorg.2023.107020] [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: 09/19/2023] [Revised: 12/01/2023] [Accepted: 12/04/2023] [Indexed: 01/06/2024]
Abstract
Abnormally high levels of copper in tumors stimulate malignant proliferation and migration of cancer cells, which proposes a formidable challenge for the thorough therapy of malignant tumors. In this work, we developed a reliable, mitochondria-targeted near-infrared aggregation-induced emission fluorescent probe, TTQ-Th, whose thiourea moiety specifically could recognize mitochondria even both upon loss of mitochondrial membrane potential or in fixated cells, and can capture copper overexpressed by tumor cells, leading to severe copper deficiency. In parallel, TTQ-Th can generate sufficient reactive oxygen species (ROS) upon photoexcitation, while copper deficiency inhibits expression of related copper-based enzymes, resulting in a decline in ATP production. Such energy deficiency, combined with reduced MMP and elevated oxidative stress can lead to critical cell oncosis. Both in vitro and intracellular experiments can illustrate that the elevated ROS has remarkable damage to tumor cells and contributes to the elimination of the primary tumor, while copper deficiency further hinder tumor cell migration and induces G0/G1 cell cycle arrest in a dose-dependent manner, which is an efficacious strategy for the treatment of malignant tumors.
Collapse
Affiliation(s)
- Ke Xue
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Yongfei Zhao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Saidong Sun
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Yuanhang Li
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China
| | - Zhengjian Qi
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, Jiangsu 211189, PR China.
| |
Collapse
|
16
|
Gong H, Zhong H, Cheng L, Li LP, Zhang DK. Post-translational protein lactylation modification in health and diseases: a double-edged sword. J Transl Med 2024; 22:41. [PMID: 38200523 PMCID: PMC10777551 DOI: 10.1186/s12967-023-04842-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
As more is learned about lactate, it acts as both a product and a substrate and functions as a shuttle system between different cell populations to provide the energy for sustaining tumor growth and proliferation. Recent discoveries of protein lactylation modification mediated by lactate play an increasingly significant role in human health (e.g., neural and osteogenic differentiation and maturation) and diseases (e.g., tumors, fibrosis and inflammation, etc.). These views are critically significant and first described in detail in this review. Hence, here, we focused on a new target, protein lactylation, which may be a "double-edged sword" of human health and diseases. The main purpose of this review was to describe how protein lactylation acts in multiple physiological and pathological processes and their potential mechanisms through an in-depth summary of preclinical in vitro and in vivo studies. Our work aims to provide new ideas for treating different diseases and accelerate translation from bench to bedside.
Collapse
Affiliation(s)
- Hang Gong
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Huang Zhong
- Department of Gastroenterology, Zigong First People's Hospital, Zigong, Sichuan, China
| | - Long Cheng
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Liang-Ping Li
- Department of Gastroenterology, Sichuan Academy of Medical Sciences and Sichuan People's Hospital, Chengdu, Sichuan, China.
| | - De-Kui Zhang
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
17
|
Moreddu R. Nanotechnology and Cancer Bioelectricity: Bridging the Gap Between Biology and Translational Medicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304110. [PMID: 37984883 PMCID: PMC10767462 DOI: 10.1002/advs.202304110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/25/2023] [Indexed: 11/22/2023]
Abstract
Bioelectricity is the electrical activity that occurs within living cells and tissues. This activity is critical for regulating homeostatic cellular function and communication, and disruptions of the same can lead to a variety of conditions, including cancer. Cancer cells are known to exhibit abnormal electrical properties compared to their healthy counterparts, and this has driven researchers to investigate the potential of harnessing bioelectricity as a tool in cancer diagnosis, prognosis, and treatment. In parallel, bioelectricity represents one of the means to gain fundamental insights on how electrical signals and charges play a role in cancer insurgence, growth, and progression. This review provides a comprehensive analysis of the literature in this field, addressing the fundamentals of bioelectricity in single cancer cells, cancer cell cohorts, and cancerous tissues. The emerging role of bioelectricity in cancer proliferation and metastasis is introduced. Based on the acknowledgement that this biological information is still hard to access due to the existing gap between biological findings and translational medicine, the latest advancements in the field of nanotechnologies for cellular electrophysiology are examined, as well as the most recent developments in micro- and nano-devices for cancer diagnostics and therapy targeting bioelectricity.
Collapse
|
18
|
Mansour N, Mehanna S, Bodman-Smith K, Daher CF, Khnayzer RS. A Ru(II)-Strained Complex with 2,9-Diphenyl-1,10-phenanthroline Ligand Induces Selective Photoactivatable Chemotherapeutic Activity on Human Alveolar Carcinoma Cells via Apoptosis. Pharmaceuticals (Basel) 2023; 17:50. [PMID: 38256884 PMCID: PMC10819265 DOI: 10.3390/ph17010050] [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: 10/25/2023] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 01/24/2024] Open
Abstract
[Ru(bipy)2(dpphen)]Cl2 (where bipy = 2,2'-bipyridine and dpphen = 2,9-diphenyl-1,10-phenanthroline) (complex 1) is a sterically strained compound that exhibits promising in vitro photocytotoxicity on an array of cell lines. Since lung adenocarcinoma cancer remains the most common lung cancer and the leading cause of cancer deaths, the current study aims to evaluate the plausible effect and uptake of complex 1 on human alveolar carcinoma cells (A549) and mesenchymal stem cells (MSC), and assess its cytotoxicity in vitro while considering its effect on cell morphology, membrane integrity and DNA damage. MSC and A549 cells showed similar rates of complex 1 uptake with a plateau at 12 h. Upon photoactivation, complex 1 exhibited selective, potent anticancer activity against A549 cells with phototoxicity index (PI) values of 16, 25 and 39 at 24, 48 and 72 h, respectively. This effect was accompanied by a significant increase in A549-cell rounding and detachment, loss of membrane integrity and DNA damage. Flow cytometry experiments confirmed that A549 cells undergo apoptosis when treated with complex 1 followed by photoactivation. In conclusion, this present study suggests that complex 1 might be a promising candidate for photochemotherapy with photoproducts that possess selective anticancer effects in vitro. These results are encouraging to probe the potential activity of this complex in vivo.
Collapse
Affiliation(s)
- Najwa Mansour
- Department of Natural Sciences, Lebanese American University, Chouran, Beirut 1102-2801, Lebanon; (N.M.); (S.M.); (C.F.D.)
| | - Stephanie Mehanna
- Department of Natural Sciences, Lebanese American University, Chouran, Beirut 1102-2801, Lebanon; (N.M.); (S.M.); (C.F.D.)
| | - Kikki Bodman-Smith
- Department of Microbial and Cellular Sciences, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, UK;
| | - Costantine F. Daher
- Department of Natural Sciences, Lebanese American University, Chouran, Beirut 1102-2801, Lebanon; (N.M.); (S.M.); (C.F.D.)
| | - Rony S. Khnayzer
- Department of Natural Sciences, Lebanese American University, Chouran, Beirut 1102-2801, Lebanon; (N.M.); (S.M.); (C.F.D.)
| |
Collapse
|
19
|
Zhan Q, Liu B, Situ X, Luo Y, Fu T, Wang Y, Xie Z, Ren L, Zhu Y, He W, Ke Z. New insights into the correlations between circulating tumor cells and target organ metastasis. Signal Transduct Target Ther 2023; 8:465. [PMID: 38129401 PMCID: PMC10739776 DOI: 10.1038/s41392-023-01725-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Organ-specific metastasis is the primary cause of cancer patient death. The distant metastasis of tumor cells to specific organs depends on both the intrinsic characteristics of the tumor cells and extrinsic factors in their microenvironment. During an intermediate stage of metastasis, circulating tumor cells (CTCs) are released into the bloodstream from primary and metastatic tumors. CTCs harboring aggressive or metastatic features can extravasate to remote sites for continuous colonizing growth, leading to further lesions. In the past decade, numerous studies demonstrated that CTCs exhibited huge clinical value including predicting distant metastasis, assessing prognosis and monitoring treatment response et al. Furthermore, increasingly numerous experiments are dedicated to identifying the key molecules on or inside CTCs and exploring how they mediate CTC-related organ-specific metastasis. Based on the above molecules, more and more inhibitors are being developed to target CTCs and being utilized to completely clean CTCs, which should provide promising prospects to administer advanced tumor. Recently, the application of various nanomaterials and microfluidic technologies in CTCs enrichment technology has assisted to improve our deep insights into the phenotypic characteristics and biological functions of CTCs as a potential therapy target, which may pave the way for us to make practical clinical strategies. In the present review, we mainly focus on the role of CTCs being involved in targeted organ metastasis, especially the latest molecular mechanism research and clinical intervention strategies related to CTCs.
Collapse
Affiliation(s)
- Qinru Zhan
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Bixia Liu
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Xiaohua Situ
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Yuting Luo
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Tongze Fu
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Yanxia Wang
- Zhongshan School of Medicine, Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Zhongpeng Xie
- Zhongshan School of Medicine, Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Lijuan Ren
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China
| | - Ying Zhu
- Department of Radiology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China.
| | - Weiling He
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY, 10065, USA.
- School of Medicine, Xiang'an Hospital of Xiamen University, Xiamen University, 361000, Xiamen, Fujian, P.R. China.
| | - Zunfu Ke
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China.
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510000, Guangzhou, Guangdong, P.R. China.
| |
Collapse
|
20
|
Tanwar S, Ghaemi B, Raj P, Singh A, Wu L, Yuan Y, Arifin DR, McMahon MT, Bulte JWM, Barman I. A Smart Intracellular Self-Assembling Bioorthogonal Raman Active Nanoprobe for Targeted Tumor Imaging. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304164. [PMID: 37715297 PMCID: PMC10700673 DOI: 10.1002/advs.202304164] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/21/2023] [Indexed: 09/17/2023]
Abstract
Inspired by the principle of in situ self-assembly, the development of enzyme-activated molecular nanoprobes can have a profound impact on targeted tumor detection. However, despite their intrinsic promise, obtaining an optical readout of enzyme activity with high specificity in native milieu has proven to be challenging. Here, a fundamentally new class of Raman-active self-assembling bioorthogonal enzyme recognition (nanoSABER) probes for targeted tumor imaging is reported. This class of Raman probes presents narrow spectral bands reflecting their vibrational fingerprints and offers an attractive solution for optical imaging at different bio-organization levels. The optical beacon harnesses an enzyme-responsive peptide sequence, unique tumor-penetrating properties, and vibrational tags with stretching frequencies in the cell-silent Raman window. The design of nanoSABER is tailored and engineered to transform into a supramolecular structure exhibiting distinct vibrational signatures in presence of target enzyme, creating a direct causality between enzyme activity and Raman signal. Through the integration of substrate-specific for tumor-associated enzyme legumain, unique capabilities of nanoSABER for imaging enzyme activity at molecular, cellular, and tissue levels in combination with machine learning models are shown. These results demonstrate that the nanoSABER probe may serve as a versatile platform for Raman-based recognition of tumor aggressiveness, drug accumulation, and therapeutic response.
Collapse
Affiliation(s)
- Swati Tanwar
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Behnaz Ghaemi
- The Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
- Cellular Imaging Section and Vascular Biology ProgramInstitute for Cell EngineeringThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Piyush Raj
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Aruna Singh
- The Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
- F.M. Kirby Research Center for Functional Brain ImagingKennedy Krieger Inc.BaltimoreMD21205USA
| | - Lintong Wu
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Yue Yuan
- Department of ChemistryUniversity of Science and Technology of China96 Jinzhai RoadHefeiAnhui230026China
| | - Dian R. Arifin
- The Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
- Cellular Imaging Section and Vascular Biology ProgramInstitute for Cell EngineeringThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
| | - Michael T. McMahon
- The Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
- F.M. Kirby Research Center for Functional Brain ImagingKennedy Krieger Inc.BaltimoreMD21205USA
| | - Jeff W. M. Bulte
- The Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
- Cellular Imaging Section and Vascular Biology ProgramInstitute for Cell EngineeringThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
- F.M. Kirby Research Center for Functional Brain ImagingKennedy Krieger Inc.BaltimoreMD21205USA
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Department of Chemical & Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Department of OncologyJohns Hopkins UniversityBaltimoreMD21231USA
| | - Ishan Barman
- Department of Mechanical EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- The Russell H. Morgan Department of Radiology and Radiological ScienceThe Johns Hopkins University School of MedicineBaltimoreMD21205USA
- Department of OncologyJohns Hopkins UniversityBaltimoreMD21231USA
| |
Collapse
|
21
|
Amer L, Retout M, Jokerst JV. Activatable prodrug for controlled release of an antimicrobial peptide via the proteases overexpressed in Candida albicans and Porphyromonas gingivalis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.27.568833. [PMID: 38076788 PMCID: PMC10705279 DOI: 10.1101/2023.11.27.568833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
We report the controlled release of an antimicrobial peptide using enzyme-activatable prodrugs to treat and detect Candida albicans and Porphyromonas gingivalis . Our motivation lies in the prevalence of these microorganisms in the subgingival area where the frequency of fungal colonization increases with periodontal disease. This work is based on an antimicrobial peptide that is both therapeutic and induces a color change in a nanoparticle reporter. This antimicrobial peptide was then built into a zwitterionic prodrug that quenches its activity until activation by a protease inherent to these pathogens of interest: SAP9 or RgpB for C. albicans and P. gingivalis , respectively. We first confirmed that the intact zwitterionic prodrug has negligible toxicity to fungal, bacterial, and mammalian cells absent a protease trigger. Next, the therapeutic impact was assessed via disk diffusion and viability assays and showed a minimum inhibitory concentration of 3.1 - 16 µg/mL, which is comparable to the antimicrobial peptide alone (absent integration into prodrug). Finally, the zwitterionic design was exploited for colorimetric detection of C. albicans and P. gingivalis proteases. When the prodrugs were cleaved, the plasmonic nanoparticles aggregated causing a color change with a limit of detection of 10 nM with gold nanoparticles and 3 nM with silver nanoparticles. This approach has value as a convenient and selective protease sensing and protease-induced treatment mechanism based on bioinspired antimicrobial peptides. Abstract Figure
Collapse
|
22
|
Mondal T, Gaur H, Wamba BEN, Michalak AG, Stout C, Watson MR, Aleixo SL, Singh A, Condello S, Faller R, Leiserowitz GS, Bhatnagar S, Tushir-Singh J. Characterizing the regulatory Fas (CD95) epitope critical for agonist antibody targeting and CAR-T bystander function in ovarian cancer. Cell Death Differ 2023; 30:2408-2431. [PMID: 37838774 PMCID: PMC10657439 DOI: 10.1038/s41418-023-01229-7] [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: 05/24/2023] [Revised: 09/14/2023] [Accepted: 09/28/2023] [Indexed: 10/16/2023] Open
Abstract
Receptor clustering is the most critical step to activate extrinsic apoptosis by death receptors belonging to the TNF superfamily. Although clinically unsuccessful, using agonist antibodies, the death receptors-5 remains extensively studied from a cancer therapeutics perspective. However, despite its regulatory role and elevated function in ovarian and other solid tumors, another tumor-enriched death receptor called Fas (CD95) remained undervalued in cancer immunotherapy until recently, when its role in off-target tumor killing by CAR-T therapies was imperative. By comprehensively analyzing structure studies in the context of the binding epitope of FasL and various preclinical Fas agonist antibodies, we characterize a highly significant patch of positively charged residue epitope (PPCR) in its cysteine-rich domain 2 of Fas. PPCR engagement is indispensable for superior Fas agonist signaling and CAR-T bystander function in ovarian tumor models. A single-point mutation in FasL or Fas that interferes with the PPCR engagement inhibited apoptotic signaling in tumor cells and T cells. Furthermore, considering that clinical and immunological features of the autoimmune lymphoproliferative syndrome (ALPS) are directly attributed to homozygous mutations in FasL, we reveal differential mechanistic details of FasL/Fas clustering at the PPCR interface compared to described ALPS mutations. As Fas-mediated bystander killing remains vital to the success of CAR-T therapies in tumors, our findings highlight the therapeutic analytical design for potentially effective Fas-targeting strategies using death agonism to improve cancer immunotherapy in ovarian and other solid tumors.
Collapse
Affiliation(s)
- Tanmoy Mondal
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Himanshu Gaur
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Brice E N Wamba
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Abby Grace Michalak
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- Undergraduate Research Program Volunteers, University of California Davis, Davis, CA, USA
| | - Camryn Stout
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- Undergraduate Research Program Volunteers, University of California Davis, Davis, CA, USA
| | - Matthew R Watson
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- Undergraduate Research Program Volunteers, University of California Davis, Davis, CA, USA
| | - Sophia L Aleixo
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- Undergraduate Research Program Volunteers, University of California Davis, Davis, CA, USA
| | - Arjun Singh
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
| | - Salvatore Condello
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Roland Faller
- Department of Chemical Engineering, University of California Davis, Davis, CA, USA
| | - Gary Scott Leiserowitz
- Department of Obstetrics and Gynecology, UC Davis School of Medicine, Sacramento, CA, USA
- UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - Sanchita Bhatnagar
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA
- UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA
| | - Jogender Tushir-Singh
- Laboratory of Novel Biologics, University of California Davis, Davis, CA, USA.
- Department of Medical Microbiology and Immunology, University of California Davis, Davis, CA, USA.
- UC Davis Comprehensive Cancer Center, UC Davis School of Medicine, Sacramento, CA, USA.
- Ovarian Cancer Academy Early Career Investigator at UC Davis, Davis, CA, USA.
| |
Collapse
|
23
|
Xu R, Wang Q, Zhu J, Bei Y, Chu Y, Sun Z, Du S, Zhou S, Ding N, Meng F, Liu B. Membrane fusogenic nanoparticle-based HLA-peptide-addressing universal T cell receptor-engineered T (HAUL TCR-T) cell therapy in solid tumor. Bioeng Transl Med 2023; 8:e10585. [PMID: 38023696 PMCID: PMC10658479 DOI: 10.1002/btm2.10585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 12/01/2023] Open
Abstract
T cell receptor-engineered T (TCR-T) cell therapy has demonstrated therapeutic effects in basic research and clinical trials for treating solid tumors. Due to the peptide-dependent recognition and the human leukocyte antigen (HLA)-restriction, TCR-T cell therapy is generally custom designed to target individual antigens. The lack of suitable universal targets for tumor cells significantly limits its clinical applications. Establishing a universal TCR-T treatment strategy is of great significance. This study designed and evaluated the HLA-peptide-addressing universal (HAUL) TCR-T cell therapy based on HLA-peptide (pHLA) loaded membrance fusogenic deliver system. The pHLA-NP-based tumor cell membrane modification technology can transfer the pHLA onto the surface of tumor cells through membrane fusogenic nanoparticles. Then tumor cells are recognized and killed by TCR-T cells specifically. The HAUL TCR-T cell therapy technology is a universal technology that enables tumor cells to be identified and killed by specific TCR-T cells, regardless of the HLA typing of tumor cells.
Collapse
Affiliation(s)
- Ruihan Xu
- The Comprehensive Cancer Centre of Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Qin Wang
- The Comprehensive Cancer Centre of Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Junmeng Zhu
- The Comprehensive Cancer Centre of Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Yuncheng Bei
- The Comprehensive Cancer Centre of Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Yanhong Chu
- The Comprehensive Cancer Centre of Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Zhichen Sun
- The Comprehensive Cancer Centre of Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Shiyao Du
- The Comprehensive Cancer Centre of Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Shujuan Zhou
- The Comprehensive Cancer Centre of Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Naiqing Ding
- The Comprehensive Cancer Centre of Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Fanyan Meng
- The Comprehensive Cancer Centre of Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| | - Baorui Liu
- The Comprehensive Cancer Centre of Nanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjingChina
| |
Collapse
|
24
|
Dong J, Zheng Z, Zhou M, Wang Y, Chen J, Cen J, Cao T, Yang T, Xu Y, Shu G, Lu X, Liang Y. EGCG-LYS Fibrils-Mediated CircMAP2K2 Silencing Decreases the Proliferation and Metastasis Ability of Gastric Cancer Cells in Vitro and in Vivo. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304075. [PMID: 37752765 PMCID: PMC10646246 DOI: 10.1002/advs.202304075] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/04/2023] [Indexed: 09/28/2023]
Abstract
Aberrant expression of circular RNAs (circRNAs) has been reported to play an important biological regulatory role in gastric cancer (GC). For the purpose of silencing cancer-related genes, a new approach for cancer treatment using nanocarriers to deliver siRNA has been proposed. In this study, abundantly expressed circMAP2K2 (hsa_circRNA_102415) is identified in GC cells. CircMAP2K2 regulates the PCBP1/GPX1 axis through proteasome-mediated degradation, which further mediates the activation of the AKT/GSK3β/epithelial-to-mesenchymal transition (EMT) signaling pathway and enhances the proliferation and metastatic ability of GC cells. To establish novel GC treatment, epigallocatechin-3-gallate-lysozyme (EGCG-LYS) fibrils are synthesized, and in vitro experiments demonstrate that EGCG-LYS has a higher siRNA delivery efficiency than Lipofectamine 2000 (lipo2000), which effectively silences the expression of circMAP2K2. Further studies show that EGCG-LYS carrying siRNA can successfully achieve lysosome escape, which allows it to be located in the cytoplasm to achieve post-transcriptional gene silencing. In addition, EGCG-LYS carrying si-circMAP2K2 has good circulating stability, excellent biosafety and antitumor ability in vivo. The EGCG-LYS fibrils delivery system provides a new tool and approach for the treatment of GC.
Collapse
Affiliation(s)
- Jiaqi Dong
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan Road IIGuangzhou510080P. R. China
- The 10th Affiliated Hospital of Southern Medical University (Dongguan People's Hospital)Southern Medical UniversityNo. 78, Wandao RoadDongguan523059P. R. China
| | - Zhousan Zheng
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan Road IIGuangzhou510080P. R. China
- State Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterNo. 651, Dongfeng East RoadGuangzhou510060P. R. China
| | - Mi Zhou
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan Road IIGuangzhou510080P. R. China
| | - Yunfei Wang
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan Road IIGuangzhou510080P. R. China
| | - Jiajie Chen
- Department of PediatricsThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan Road IIGuangzhou510080P. R. China
| | - Junjie Cen
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan Road IIGuangzhou510080P. R. China
| | - Tiefeng Cao
- Department of GynecologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan Road IIGuangzhou510080P. R. China
| | - Taowei Yang
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan Road IIGuangzhou510080P. R. China
| | - Yi Xu
- Department of OncologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan Road IIGuangzhou510080P. R. China
| | - Guannan Shu
- Department of UrologyThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan Road IIGuangzhou510080P. R. China
| | - Xuanxuan Lu
- Department of Food Science and EngineeringJinan UniversityNo. 601, West Huangpu AvenueGuangzhou510632P. R. China
| | - Yanping Liang
- Department of Laboratory MedicineThe First Affiliated Hospital of Sun Yat‐sen UniversityNo. 58, Zhongshan Road IIGuangzhou510080P. R. China
| |
Collapse
|
25
|
Wu Y, Chen X, Zhu L, Wang D, Li X, Song J, Wang D, Yu X, Li Y, Tang BZ. Endoplasmic Reticulum-Targeted Aggregation-Induced Emission Luminogen for Synergetic Tumor Ablation with Glibenclamide. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37903083 DOI: 10.1021/acsami.3c10940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2023]
Abstract
Photodynamic therapy based on fluorescence illumination of subcellular organelles and in situ bursts of reactive oxygen species (ROS) has been recognized as a promising strategy for cancer theranostics. However, the short life of ROS and unclarified anticancer mechanism seriously restrict the application. Herein, we rationally designed and facilely synthesized a 2,6-dimethylpyridine-based triphenylamine (TPA) derivative TPA-DMPy with aggregation-induced emission (AIE) features and production of type-I ROS. Except for its selective binding to the endoplasmic reticulum (ER), TPA-DMPy, in synergy with glibenclamide, a medicinal agent used against diabetes, induced significant apoptosis of cancer cells in vitro and in vivo. Additionally, TPA-DMPy greatly incited the release of calcium from ER upon light irradiation to further aggravate the depolarization of ER membrane potential caused by glibenclamide, thus inducing fatal ER stress and crosstalk between ER and mitochondria. Our study extends the biological design and application of AIE luminogens and provides new insights into discovering novel anticancer targets and agents.
Collapse
Affiliation(s)
- Yifan Wu
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Xiaohui Chen
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
- Institute of Laboratory Medicine, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, School of Medical Technology, Guangdong Medical University, Dongguan, Guangdong 523808, China
| | - Liwei Zhu
- Innovation Research Center for AIE Pharmaceutical Biology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Deliang Wang
- Department of Materials Chemistry, Huzhou University, Huzhou, Zhejiang 313000, China
| | - Xue Li
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Jiayi Song
- Innovation Research Center for AIE Pharmaceutical Biology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Dong Wang
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
| | - Xiyong Yu
- Innovation Research Center for AIE Pharmaceutical Biology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Ying Li
- Center for AIE Research, Shenzhen Key Laboratory of Polymer Science and Technology, Guangdong Research Center for Interfacial Engineering of Functional Materials, College of Materials Science and Engineering, Shenzhen University, Shenzhen, Guangdong 518060, China
- Innovation Research Center for AIE Pharmaceutical Biology, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Ben Zhong Tang
- Shenzhen Institute of Aggregate Science and Engineering, School of Science and Engineering, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| |
Collapse
|
26
|
Zandi A, Shojaeian F, Abbasvandi F, Faranoush M, Anbiaee R, Hoseinpour P, Gilani A, Saghafi M, Zandi A, Hoseinyazdi M, Davari Z, Miraghaie SH, Tayebi M, Taheri MS, Ardestani SMS, Sheikhi Mobarakeh Z, Nikshoar MR, Enjavi MH, Kordehlachin Y, Mousavi-kiasary SMS, Mamdouh A, Akbari ME, Yunesian M, Abdolahad M. A human pilot study on positive electrostatic charge effects in solid tumors of the late-stage metastatic patients. Front Med (Lausanne) 2023; 10:1195026. [PMID: 37915327 PMCID: PMC10616960 DOI: 10.3389/fmed.2023.1195026] [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: 03/27/2023] [Accepted: 09/25/2023] [Indexed: 11/03/2023] Open
Abstract
Background Correlative interactions between electrical charges and cancer cells involve important unknown factors in cancer diagnosis and treatment. We previously reported the intrinsic suppressive effects of pure positive electrostatic charges (PEC) on the proliferation and metabolism of invasive cancer cells without any effect on normal cells in cell lines and animal models. The proposed mechanism was the suppression of pro-caspases 3 and 9 with an increase in Bax/Bcl2 ratio in exposed malignant cells and perturbation induced in the KRAS pathway of malignant cells by electrostatic charges due to the phosphate molecule electrostatic charge as the trigger of the pathway. This study aimed to examine PECs as a complementary treatment for patients with different types of solid metastatic tumors, who showed resistance to chemotherapy and radiotherapy. Methods In this study, solid metastatic tumors of the end-stage patients (n = 41) with various types of cancers were locally exposed to PEC for at least one course of 12 days. The patient's signs and symptoms, the changes in their tumor size, and serum markers were followed up from 30 days before positive electrostatic charge treating (PECT) until 6 months after the study. Results Entirely, 36 patients completed the related follow-ups. Significant reduction in tumor sizes and cancer-associated enzymes as well as improvement in cancer-related signs and symptoms and patients' lifestyles, without any side effects on other tissues or metabolisms of the body, were observed in more than 80% of the candidates. Conclusion PECT induced significant cancer remission in combination with other therapies. Therefore, this non-ionizing radiation would be a beneficial complementary therapy, with no observable side effects of ionizing radiotherapy, such as post-radiation inflammation.
Collapse
Affiliation(s)
- Ashkan Zandi
- Nano Electronic Centre of Excellence, Nanobioelectronic Devices Laboratory, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
- Nano Electronic Centre of Excellence, Nanoelectronics and Thin Film Laboratory, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | - Fatemeh Shojaeian
- Nano Electronic Centre of Excellence, Nanobioelectronic Devices Laboratory, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fereshteh Abbasvandi
- Department of ATMP, Breast Cancer Research Centre, Motamed Cancer Institute, ACECR, Tehran, Iran
- Cancer Research Centre, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Faranoush
- Pediatric Growth and Development Research Centre, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
- Cardio-Oncology Research Centre, Rajaie Cardiovascular Medical and Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Robab Anbiaee
- Department of Radiation Oncology, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parisa Hoseinpour
- Nano Electronic Centre of Excellence, Nanobioelectronic Devices Laboratory, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
- SEPAS Pathology Laboratory, Tehran, Iran
| | - Ali Gilani
- Nano Electronic Centre of Excellence, Nanobioelectronic Devices Laboratory, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | - Mohammad Saghafi
- Nano Electronic Centre of Excellence, Nanobioelectronic Devices Laboratory, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | - Afsoon Zandi
- Department of Otolaryngology, Head and Neck Surgery, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meisam Hoseinyazdi
- Medical Imaging Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Davari
- Nano Electronic Centre of Excellence, Nanobioelectronic Devices Laboratory, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | - Seyyed Hossein Miraghaie
- Nano Electronic Centre of Excellence, Nanobioelectronic Devices Laboratory, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | - Mahtab Tayebi
- Department of ATMP, Breast Cancer Research Centre, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Morteza Sanei Taheri
- Department of Radiology, Shohada Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - S. Mehdi Samimi Ardestani
- Department of Psychiatry, Behavioural Sciences Research Centre, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Sheikhi Mobarakeh
- Department of Quality of Life, Breast Cancer Research Centre, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Reza Nikshoar
- Department of Gastroenterology Surgery, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hossein Enjavi
- Nano Electronic Centre of Excellence, Nanobioelectronic Devices Laboratory, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
- Nano Electronic Centre of Excellence, Nanoelectronics and Thin Film Laboratory, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | - Yasin Kordehlachin
- Nano Electronic Centre of Excellence, Nanobioelectronic Devices Laboratory, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | - S. M. Sadegh Mousavi-kiasary
- Nano Electronic Centre of Excellence, Nanobioelectronic Devices Laboratory, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | - Amir Mamdouh
- Nano Electronic Centre of Excellence, Nanobioelectronic Devices Laboratory, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
| | | | - Masud Yunesian
- Department of Environmental Health Engineering, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- Department of Research Methodology and Data Analysis, Institute for Environmental Research, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdolahad
- Nano Electronic Centre of Excellence, Nanobioelectronic Devices Laboratory, Cancer Electronics Research Group, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
- Nano Electronic Centre of Excellence, Nanoelectronics and Thin Film Laboratory, School of Electrical and Computer Engineering, Faculty of Engineering, University of Tehran, Tehran, Iran
- Imam-Khomeini Hospital, Tehran University of Medical Sciences, Cancer Institute, Tehran, Iran
- UT&TUMS Cancer Electrotechnique Research Centre, YAS Hospital, Tehran, Iran
| |
Collapse
|
27
|
Furukawa N, Yang W, Chao A, Patil A, Mirando A, Pandey N, Popel A. Chemokine-derived oncolytic peptide induces immunogenic cancer cell death and significantly suppresses tumor growth. RESEARCH SQUARE 2023:rs.3.rs-3335225. [PMID: 37886580 PMCID: PMC10602061 DOI: 10.21203/rs.3.rs-3335225/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Chemokinostatin-1 (CKS1) is a 24-mer peptide originally discovered as an anti-angiogenic peptide derived from the CXCL1 chemokine. Here, we demonstrate that CKS1 acts not only as an anti-angiogenic peptide but also as an oncolytic peptide due to its structural and physical properties. CKS1 induced both necrotic and apoptotic cell death specifically in cancer cells while showing minimal toxicity in non-cancerous cells. Mechanistically, CKS1 disrupted the cell membrane of cancer cells quickly after treatment and activated the apoptotic pathway at later time points. Furthermore, immunogenic molecules were released from CKS1 treated cells, indicating that CKS1 induces immunogenic cell death. CKS1 effectively suppressed tumor growth in vivo. Collectively, these data demonstrate that CKS1 is a unique peptide that functions both as an anti-angiogenic peptide and as an oncolytic peptide and has a therapeutic potential to treat cancer.
Collapse
Affiliation(s)
| | - Wendy Yang
- Johns Hopkins University School of Medicine
| | - Alex Chao
- Johns Hopkins University School of Medicine
| | | | | | | | | |
Collapse
|
28
|
Yan H, Xu P, Ma H, Li Y, Zhang R, Cong H, Yu B, Shen Y. Enzyme-triggered transcytosis of drug carrier system for deep penetration into hepatoma tumors. Biomaterials 2023; 301:122213. [PMID: 37385137 DOI: 10.1016/j.biomaterials.2023.122213] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 06/08/2023] [Accepted: 06/20/2023] [Indexed: 07/01/2023]
Abstract
In recent years, nano-drug delivery systems have made considerable progress in the direction of tumor treatment, but the low permeability of drugs has restricted the development of nano drugs. To solve this problem, we constructed a nano-drug delivery system with the dual effects of γ-glutamyltransferase (GGT) reaction and high nuclear targeting in tumor microenvironment to promote the deep penetration of drugs. Over-expression of GGT in tumor cells can specifically recognize γ-glutamyl substrate and release amino group from the hydrolysis reaction, which makes the whole system change from negative or neutral to positive charge system. The conjugated complex with positive charge rapidly endocytosis through electrostatic interaction, enhancing its permeability in tumor parenchyma. At the same time, the cell penetrating TAT contains a large amount of lysine, which can be identified by the nuclear pore complexes (NPCs) on the surface of the nuclear membrane, showing excellent nuclear localization function. The active DOX is released in the nucleus, which inhibits the mitosis of cancer cells and enhances the active transport ability of drugs in tumor cells. Therefore, this drug delivery system actively transports adriamycin into the tumor to achieve deep penetration of drugs through enzyme response and nuclear targeting, showing high anti-tumor activity and can be effectively applied to the treatment of liver cancer.
Collapse
Affiliation(s)
- Han Yan
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Pengchao Xu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - He Ma
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Yanan Li
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Runfeng Zhang
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China
| | - Hailin Cong
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China; School of Materials Science and Engineering, Shandong University of Technology, Zibo 255000, China.
| | - Bing Yu
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China; State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao 266071, China.
| | - Youqing Shen
- College of Chemistry and Chemical Engineering, College of Materials Science and Engineering, Institute of Biomedical Materials and Engineering, Qingdao University, Qingdao, 266071, China; Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, And Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| |
Collapse
|
29
|
Go EJ, Yang H, Park W, Lee SJ, Han JH, Kong SJ, Lee WS, Han DK, Chon HJ, Kim C. Systemic Delivery of a STING Agonist-Loaded Positively Charged Liposome Selectively Targets Tumor Immune Microenvironment and Suppresses Tumor Angiogenesis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300544. [PMID: 37381624 DOI: 10.1002/smll.202300544] [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: 01/18/2023] [Revised: 06/13/2023] [Indexed: 06/30/2023]
Abstract
Although stimulator of interferon genes (STING) agonists has shown great promise in preclinical studies, the clinical development of STING agonist therapy is challenged by its limited systemic delivery. Here, positively charged fusogenic liposomes loaded with a STING agonist (PoSTING) are designed for systemic delivery and to preferentially target the tumor microenvironment. When PoSTING is administered intravenously, it selectively targets not only tumor cells but also immune and tumor endothelial cells (ECs). In particular, delivery of STING agonists to tumor ECs normalizes abnormal tumor vasculatures, induces intratumoral STING activation, and elicits robust anti-tumor T cell immunity within the tumor microenvironment. Therefore, PoSTING can be used as a systemic delivery platform to overcome the limitations of using STING agonists in clinical trials.
Collapse
Affiliation(s)
- Eun-Jin Go
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Hannah Yang
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Seung Joon Lee
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Jun-Hyeok Han
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seoburo 2066, Suwon, Gyeonggi, 16419, Republic of Korea
| | - So Jung Kong
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Won Suk Lee
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Hong Jae Chon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| | - Chan Kim
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
- Laboratory of Translational Immuno-Oncology, CHA University School of Medicine, Seongnam, Gyeonggi, 13496, Republic of Korea
| |
Collapse
|
30
|
Mujyambere B, Mohanakrishnan S, Jabeen Mubarak S, Vedagiri H, Ramasamy S, Samiappan S. Design, synthesis and analysis of charged RGD derivatives. Bioinformation 2023; 19:918-924. [PMID: 37928495 PMCID: PMC10625369 DOI: 10.6026/97320630019918] [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: 09/01/2023] [Revised: 09/30/2023] [Accepted: 09/30/2023] [Indexed: 11/07/2023] Open
Abstract
In the present study, negatively charged N-Biotin-RGD and positively charged C-Biotin-RGD were designed, synthesized, and characterized with docking analysis. The fixation of MDA-MB-231 cells with formalin made their cell surface neutrally charged thus removing the electrostatic interactions between charged biotinylated RGD derivatives and MDA-MB-231 cells. The results of the binding affinity of biotinylated RGD derivatives against MDA-MB-231 cells showed that N-Biotin-RGD had higher binding affinity than C-Biotin-RGD. The cytotoxic effect was analyzed by incubating charged biotinylated RGD derivatives with live MDA-MB-231 cells. MDA-MB-231 cell surface is negatively charged due to high hypersialyliation of polyglycans and Warburg effect. The results of their cytotoxic activities against live MDA-MB-231 cells were found to be electrostatic in nature. C-Biotin-RGD had an attractive interaction with the MDA-MB-231 cell surface resulting in a higher cytotoxic effect. In comparison, N-Biotin-RGD had a repulsive interaction with the MDA-MB-231 cell surface resulting in a lower cytotoxic effect. Hence, positively charged C-Biotin-RGD is a better cytotoxic agent than a negatively charged N-Biotin-RGD against MDA-MB-231 cells.
Collapse
Affiliation(s)
| | | | | | - Hemamalini Vedagiri
- Department of Bioinformatics, Bharathiar University, Coimbatore, Tamilnadu, India
| | - Sivasamy Ramasamy
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamilnadu, India
| | - Suja Samiappan
- Department of Bioinformatics, Bharathiar University, Coimbatore, Tamilnadu, India
| |
Collapse
|
31
|
Samanta S, Mahapatra P, Ohshima H, Gopmandal PP. Diffusiophoresis of Weakly Charged Fluid Droplets in a General Electrolyte Solution: An Analytical Theory. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:12452-12466. [PMID: 37615654 DOI: 10.1021/acs.langmuir.3c01667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Owing to the importance of analytical results for electrokinetics of colloidal entities, we performed a mathematical analysis to determine the closed form analytical results for the diffusiophoretic velocity of a hydrophobic and polarizable fluid droplet. A comprehensive mathematical model is developed for diffusiophoresis, considering the background aqueous medium as general electrolytes (e.g., binary valence-symmetric/asymmetric electrolytes and a mixed solution of binary electrolytes). We performed our analysis under a weak concentration gradient, and the analytical results for diffusiophoretic velocity are calculated within the Debye-Hückel electrostatic framework. The exact form of the diffusiophoretic velocity is further approximated with negligible error, and the approximate form is found to be free from any of the cumbersome exponential integrals and thus very convenient for practical use. The present theory also covers the diffusiophoresis of perfectly dielectric as well as perfectly conducting droplets as its limiting case. In addition, we have further derived a number of closed form expressions for diffusiophoretic velocity pertaining to several particular cases, and we observed that the derived limit correctly recovers the available existing analytical results for diffusiophoretic velocity. Thus, the present analytical theory for diffusiophoresis can be applied to a wide class of fluidic droplets, e.g., hydrophobic and dielectric oil/conducting mercury droplets, air bubbles, nanoemulsions, as well as any polarizable and hydrophobic fluidic droplet suspended in a solution of general electrolytes.
Collapse
Affiliation(s)
- Susmita Samanta
- Department of Mathematics, National Institute of Technology Durgapur, Durgapur 713209, India
| | - Paramita Mahapatra
- Department of Mathematics, National Institute of Technology Durgapur, Durgapur 713209, India
| | - H Ohshima
- Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Yamazaki, Noda, Chiba 278-8510, Japan
| | - Partha P Gopmandal
- Department of Mathematics, National Institute of Technology Durgapur, Durgapur 713209, India
| |
Collapse
|
32
|
Iliev I, Mavrova A, Yancheva D, Dimov S, Staneva G, Nesheva A, Tsoneva I, Nikolova B. 2-Alkyl-Substituted-4-Amino-Thieno[2,3- d]Pyrimidines: Anti-Proliferative Properties to In Vitro Breast Cancer Models. Molecules 2023; 28:6347. [PMID: 37687177 PMCID: PMC10489817 DOI: 10.3390/molecules28176347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/24/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
Thienopyrimidines are structural analogs of quinazolines, and the creation of new 2-alkyl derivatives of ethyl 4-aminothienopyrimidine-6-carboxylates for the study of their anti-proliferative properties is of great pharmacological interest. Some 2-alkyl-4-amino-thieno[2,3-d]pyrimidines 2-5 were synthesized, and their cyto- and phototoxicity against BALB 3T3 cells were established by an in vitro 3T3 NRU test. The obtained results indicate that the tested compounds are not cytotoxic or phototoxic, and that they are appropriate to be studied for their anti-proliferative and anti-tumor properties. The anti-proliferative potential of the compounds was investigated on MCF-7 and MDA-MB-231 cancer cells, as well as a MCF-10A cell line (normal human mammary epithelial cells). The most toxic to MCF-7 was thienopyrimidine 3 with IC50 13.42 μg/mL (IC50 0.045 μM), followed by compound 4 (IC50 28.89 μg/mL or IC50 0.11 μM). The thienopyrimidine 4 revealed higher selectivity to MCF-7 and lower activity (IC50 367 μg/mL i.e., 1.4 μM) than compound 3 with MCF-10A cells. With respect to MDA-MB-231 cells, ester 2 manifested the highest effect with IC50 52.56 μg/mL (IC50 0.16 μM), and 2-ethyl derivative 4 revealed IC50 62.86 μg/mL (IC50 0.24 μM). It was estimated that the effect of the substances on the cell cycle progression was due to cell cycle arrest in the G2 stage for MDA-MB-231, while arrest in G1 was detected for the estrogen (ER)-positive MCF-7 cell line. The tested compound's effects on the change of the zeta potential in the tumorigenic cells utilized in this study were determined. The calculation which we performed of the physicochemical properties and pharmacokinetic parameters influencing the biological activity suggested high intestinal absorption, as well as drug-likeness.
Collapse
Affiliation(s)
- Ivan Iliev
- Institute of Experimental Morphology, Pathology and Anthropology with Museum, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 25, 1113 Sofia, Bulgaria;
| | - Anelia Mavrova
- Department of Organic Chemistry, Faculty of Chemical Technologies, University of Chemical Technology and Metallurgy, S8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria; (A.M.); (S.D.)
| | - Denitsa Yancheva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 9, 1113 Sofia, Bulgaria;
| | - Stefan Dimov
- Department of Organic Chemistry, Faculty of Chemical Technologies, University of Chemical Technology and Metallurgy, S8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria; (A.M.); (S.D.)
| | - Galya Staneva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 21, 1113 Sofia, Bulgaria; (A.N.); (I.T.)
| | - Alexandrina Nesheva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 21, 1113 Sofia, Bulgaria; (A.N.); (I.T.)
| | - Iana Tsoneva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 21, 1113 Sofia, Bulgaria; (A.N.); (I.T.)
| | - Biliana Nikolova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., bl. 21, 1113 Sofia, Bulgaria; (A.N.); (I.T.)
| |
Collapse
|
33
|
Adnan M, Akhter MH, Afzal O, Altamimi ASA, Ahmad I, Alossaimi MA, Jaremko M, Emwas AH, Haider T, Haider MF. Exploring Nanocarriers as Treatment Modalities for Skin Cancer. Molecules 2023; 28:5905. [PMID: 37570875 PMCID: PMC10421083 DOI: 10.3390/molecules28155905] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/11/2023] [Accepted: 06/16/2023] [Indexed: 08/13/2023] Open
Abstract
Cancer is a progressive disease of multi-factorial origin that has risen worldwide, probably due to changes in lifestyle, food intake, and environmental changes as some of the reasons. Skin cancer can be classified into melanomas from melanocytes and nonmelanoma skin cancer (NMSC) from the epidermally-derived cell. Together it constitutes about 95% of skin cancer. Basal cell carcinoma (BCC) and cutaneous squamous cell carcinoma (CSCC) are creditworthy of 99% of NMSC due to the limited accessibility of conventional formulations in skin cancer cells of having multiple obstacles in treatment reply to this therapeutic regime. Despite this, it often encounters erratic bioavailability and absorption to the target. Nanoparticles developed through nanotechnology platforms could be the better topical skin cancer therapy option. To improve the topical delivery, the nano-sized delivery system is appropriate as it fuses with the cutaneous layer and fluidized membrane; thus, the deeper penetration of therapeutics could be possible to reach the target spot. This review briefly outlooks the various nanoparticle preparations, i.e., liposomes, niosomes, ethosomes, transferosomes, transethosomes, nanoemulsions, and nanoparticles technologies tested into skin cancer and impede their progress tend to concentrate in the skin layers. Nanocarriers have proved that they can considerably boost medication bioavailability, lowering the frequency of dosage and reducing the toxicity associated with high doses of the medication.
Collapse
Affiliation(s)
- Mohammad Adnan
- Faculty of Pharmacy, Integral University, Lucknow 226026, Uttar Pradesh, India;
| | - Md. Habban Akhter
- School of Pharmaceutical and Population Health Informatics (SoPPHI), DIT University, Dehradun 248009, Uttarakhand, India;
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (O.A.); (A.S.A.A.); (M.A.A.)
| | - Abdulmalik S. A. Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (O.A.); (A.S.A.A.); (M.A.A.)
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 62521, Saudi Arabia;
| | - Manal A. Alossaimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia; (O.A.); (A.S.A.A.); (M.A.A.)
| | - Mariusz Jaremko
- Smart-Health Initiative (SHI) and Red Sea Research Center (RSRC), Division of Biological and Environmental Sciences and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia;
| | - Abdul-Hamid Emwas
- Core Labs, King Abdullah University of Science and Technology (KAUST), Thuwal 23955, Saudi Arabia;
| | - Tanweer Haider
- Amity Institute of Pharmacy, Amity University, Gwalior 474005, Madhya Pradesh, India;
| | - Md. Faheem Haider
- Faculty of Pharmacy, Integral University, Lucknow 226026, Uttar Pradesh, India;
| |
Collapse
|
34
|
Moharamipour S, Aminifar M, Foroughi-Gilvaee MR, Faranoush P, Mahdavi R, Abadijoo H, Parniani M, Abbasvandi F, Mansouri S, Abdolahad M. Hydroelectric actuator for 3-dimensional analysis of electrophoretic and dielectrophoretic behavior of cancer cells; suitable in diagnosis and invasion studies. BIOMATERIALS ADVANCES 2023; 151:213476. [PMID: 37276690 DOI: 10.1016/j.bioadv.2023.213476] [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: 11/13/2022] [Revised: 04/29/2023] [Accepted: 05/13/2023] [Indexed: 06/07/2023]
Abstract
Cancer is a cellular-based disease, so cytological diagnosis is one of the main challenges for its early detection. An extensive number of diagnostic methods have been developed to separate cancerous cells from normal ones, in electrical methods attract progressive attention. Identifying and specifying different cells requires understanding their dielectric and electric properties. This study evaluated MDA-MB-231, HUVEC, and MCF-10A cell lines, WBCs isolated from blood, and patient-derived cell samples with a cylindrical body with two transparent FTO (fluorine-doped tin oxide) plate electrodes. Cell mobility rates were recorded in response to these stimuli. It was observed that cancer cells demonstrate drastic changes in their motility in the presence and absence of an electric field (DC/AC). Also, solution viscosity's effect on cancer cells' capturing efficacy was evaluated. This research's main distinguished specification uses a non-microfluidic platform to detect and pathologically evaluate cytological samples with a simple, cheap, and repeatable platform. The capturing procedure was carried out on a cytological slide without any complicated electrode patterning with the ability of cytological staining. Moreover, this platform successfully designed and experimented with the invasion assay (the ability of captured cancer cells to invade normal cells).
Collapse
Affiliation(s)
- Shima Moharamipour
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mina Aminifar
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mohammad Reza Foroughi-Gilvaee
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran; Pediatric Growth and Development Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Pooya Faranoush
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran; Pediatric Growth and Development Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Reihane Mahdavi
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Hamed Abadijoo
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mohammad Parniani
- Pathology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Fereshteh Abbasvandi
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran; ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, P.O. BOX: 15179/64311, Tehran, Iran
| | - Sepideh Mansouri
- Radiation Oncology Research Center (RORC), Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdolahad
- Nano Bio Electronic Devices Lab, Cancer Electronics Research Group, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran, Iran; UT and TUMS Cancer Electronics Research Center, Tehran University of Medical Sciences, Tehran, Iran; Cancer Institute, Imam Khomeini Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
35
|
Aghaz F, Asadi Z, Sajadimajd S, Kashfi K, Arkan E, Rahimi Z. Codelivery of resveratrol melatonin utilizing pH responsive sericin based nanocarriers inhibits the proliferation of breast cancer cell line at the different pH. Sci Rep 2023; 13:11090. [PMID: 37422485 PMCID: PMC10329705 DOI: 10.1038/s41598-023-37668-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/26/2023] [Indexed: 07/10/2023] Open
Abstract
Protein-based nanocarriers have demonstrated good potential for cancer drug delivery. Silk sericin nano-particle is arguably one of the best in this field. In this study, we developed a surface charge reversal sericin-based nanocarrier to co-deliver resveratrol and melatonin (MR-SNC) to MCF-7 breast cancer cells as combination therapy. MR-SNC was fabricated with various sericin concentrations via flash-nanoprecipitation as a simple and reproducible method without complicated equipment. The nanoparticles were subsequently characterized for their size, charge, morphology and shape by dynamic light scattering (DLS) and scanning electron microscope (SEM). Nanocarriers chemical and conformational analysis were done by fourier transform infrared spectroscopy (FT-IR) and circular dichroism (CD) respectively. In vitro drug release was determined at different pH values (7.45, 6.5 and 6). The cellular uptake and cytotoxicity were studies using breast cancer MCF-7 cells. MR-SNC fabricated with the lowest sericin concentration (0.1%), showed a desirable 127 nm size, with a net negative charge at physiological pH. Sericin structure was preserved entirely in the form of nano-particles. Among the three pH values we applied, the maximum in vitro drug release was at pH 6, 6.5, and 7.4, respectively. This pH dependency showed the charge reversal property of our smart nanocarrier via changing the surface charge from negative to positive in mildly acidic pH, destructing the electrostatic interactions between sericin surface amino acids. Cell viability studies demonstrated the significant toxicity of MR-SNC in MCF-7 cells at all pH values after 48 h, suggesting a synergistic effect of combination therapy with the two antioxidants. The efficient cellular uptake of MR-SNC, DNA fragmentation and chromatin condensation was found at pH 6. Nutshell, our result indicated proficient release of the entrapped drug combination from MR-SNC in an acidic environment leading to cell apoptosis. This work introduces a smart pH-responsive nano-platform for anti-breast cancer drug delivery.
Collapse
Affiliation(s)
- Faranak Aghaz
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Asadi
- Students Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Clinical Biochemistry, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Soraya Sajadimajd
- Department of Biology, Faculty of Science, Razi University, Kermanshah, Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, USA
| | - Elham Arkan
- Nano Drug Delivery Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Zohreh Rahimi
- Department of Clinical Biochemistry, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran.
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
36
|
Wang J, Cong L, Shi W, Xu W, Xu S. Single-Cell Analysis and Classification according to Multiplexed Proteins via Microdroplet-Based Self-Driven Magnetic Surface-Enhanced Raman Spectroscopy Platforms Assisted with Machine Learning Algorithms. Anal Chem 2023. [PMID: 37419505 DOI: 10.1021/acs.analchem.3c01273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2023]
Abstract
A microdroplet-based surface-enhanced Raman spectroscopy (microdroplet SERS) platform was constructed to envelop individual cells in microdroplets, followed by the SERS detection of their extracellular vesicle-proteins (EV-proteins) via the in-drop immunoassays by use of immunomagnetic beads (iMBs) and immuno-SERS tags (iSERS tags). A unique phenomenon is found that iMBs can start a spontaneous reorientation on the probed cell surface based on the electrostatic force-driven interfacial aggregation effect, which leads EV-proteins and iSERS tags to be gathered from a liquid phase to a cell membrane interface and significantly improves SERS sensitivity to the single-cell analysis level due to the formation of numbers of SERS hotspots. Three EV-proteins from two breast cancer cell lines were collected and further analyzed by machine learning algorithmic tools, which will be helpful for a deeper understanding of breast cancer subtypes from the view of EV-proteins.
Collapse
Affiliation(s)
- Jiaqi Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Lili Cong
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Wei Shi
- Key Lab for Molecular Enzymology & Engineering of Ministry of Education, Jilin University, Changchun 130012, P. R. China
| | - Weiqing Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Institute of Theoretical Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Shuping Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- State Key Laboratory of Applied Optics, Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, P. R. China
- Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Institute of Theoretical Chemistry, Jilin University, Changchun 130012, P. R. China
| |
Collapse
|
37
|
Luo W, Bai L, Zhang J, Li Z, Liu Y, Tang X, Xia P, Xu M, Shi A, Liu X, Zhang D, Yu P. Polysaccharides-based nanocarriers enhance the anti-inflammatory effect of curcumin. Carbohydr Polym 2023; 311:120718. [PMID: 37028867 DOI: 10.1016/j.carbpol.2023.120718] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 02/11/2023] [Accepted: 02/14/2023] [Indexed: 02/27/2023]
Abstract
Curcumin (CUR) has been discovered to have many biological activities, including anti-inflammatory, anti-cancer, anti-oxygenation, anti-human immunodeficiency virus, anti-microbial and exhibits a good effect on the prevention and treatment of many diseases. However, the limited properties of CUR, including the poor solubility, bioavailability and instability caused by enzymes, light, metal irons, and oxygen, have compelled researchers to turn their attention to drug carrier application to overcome these drawbacks. Encapsulation may provide potential protective effects to the embedding materials and/or have a synergistic effect with them. Therefore, nanocarriers, especially polysaccharides-based nanocarriers, have been developed in many studies to enhance the anti-inflammatory capacity of CUR. Consequently, it's critical to review current advancements in the encapsulation of CUR using polysaccharides-based nanocarriers, as well as further study the potential mechanisms of action where polysaccharides-based CUR nanoparticles (the complex nanoparticles/Nano CUR-delivery systems) exhibit their anti-inflammatory effects. This work suggests that polysaccharides-based nanocarriers will be a thriving field in the treatment of inflammation and inflammation-related diseases.
Collapse
Affiliation(s)
- Wei Luo
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Liangyu Bai
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Zhangwang Li
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Yinuo Liu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Xiaoyi Tang
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China
| | - Panpan Xia
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang 330006, China; Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang 330006, China
| | - Minxuan Xu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang 330006, China; Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang 330006, China
| | - Ao Shi
- School of Medicine, St.George University of London, London, UK
| | - Xiao Liu
- Cardiology Department, The Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong.
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Institute for the Study of Endocrinology and Metabolism in Jiangxi Province, Nanchang 330006, China; Branch of Nationlal Clinical Research Center for Metabolic Diseases, Nanchang 330006, China.
| |
Collapse
|
38
|
Mavroidi B, Kaminari A, Sakellis E, Sideratou Z, Tsiourvas D. Carbon Dots-Biomembrane Interactions and Their Implications for Cellular Drug Delivery. Pharmaceuticals (Basel) 2023; 16:833. [PMID: 37375780 DOI: 10.3390/ph16060833] [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: 05/08/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
The effect of carbon dots (CDs) on a model blayer membrane was studied as a means of comprehending their ability to affect cell membranes. Initially, the interaction of N-doped carbon dots with a biophysical liposomal cell membrane model was investigated by dynamic light scattering, z-potential, temperature-modulated differential scanning calorimetry, and membrane permeability. CDs with a slightly positive charge interacted with the surface of the negative-charged liposomes and evidence indicated that the association of CDs with the membrane affects the structural and thermodynamic properties of the bilayer; most importantly, it enhances the bilayer's permeability against doxorubicin, a well-known anticancer drug. The results, like those of similar studies that surveyed the interaction of proteins with lipid membranes, suggest that carbon dots are partially embedded in the bilayer. In vitro experiments employing breast cancer cell lines and human healthy dermal cells corroborated the findings, as it was shown that the presence of CDs in the culture medium selectively enhanced cell internalization of doxorubicin and, subsequently, increased its cytotoxicity, acting as a drug sensitizer.
Collapse
Affiliation(s)
- Barbara Mavroidi
- Institute of Biosciences and Applications, National Centre for Scientific Research "Demokritos", 15310 Aghia Paraskevi, Greece
| | - Archontia Kaminari
- Institute of Nanoscience and Nanotechnology, National Centre for Scientific Research "Demokritos", 15310 Aghia Paraskevi, Greece
| | - Elias Sakellis
- Institute of Nanoscience and Nanotechnology, National Centre for Scientific Research "Demokritos", 15310 Aghia Paraskevi, Greece
| | - Zili Sideratou
- Institute of Nanoscience and Nanotechnology, National Centre for Scientific Research "Demokritos", 15310 Aghia Paraskevi, Greece
| | - Dimitris Tsiourvas
- Institute of Nanoscience and Nanotechnology, National Centre for Scientific Research "Demokritos", 15310 Aghia Paraskevi, Greece
| |
Collapse
|
39
|
Anticancer polypyrrole-polyethylenimine drug-free nanozyme for precise B-cell lymphoma therapy. Biomed Pharmacother 2023; 160:114397. [PMID: 36796279 DOI: 10.1016/j.biopha.2023.114397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 02/08/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
As an alternative strategy for cancer treatment, the combination of cancer nanomedicine and immunotherapy is promising with regard to efficacy and safety; however, precise modulation of the activation of antitumor immunity remains challenging. Therefore, the aim of the present study was to describe an intelligent nanocomposite polymer immunomodulator, drug-free polypyrrole-polyethyleneimine nanozyme (PPY-PEI NZ), which responds to the B-cell lymphoma tumor microenvironment, for precision cancer immunotherapy. Earlier engulfment of PPY-PEI NZs in an endocytosis-dependent manner resulted in rapid binding in four different types of B-cell lymphoma cells. The PPY-PEI NZ effectively suppressed B cell colony-like growth in vitro accompanied by cytotoxicity via apoptosis induction. During PPY-PEI NZ-induced cell death, mitochondrial swelling, loss of mitochondrial transmembrane potential (MTP), downregulation of antiapoptotic proteins, and caspase-dependent apoptosis were observed. Deregulated AKT and ERK signaling contributed to glycogen synthase kinase-3-regulated cell apoptosis following deregulation of Mcl-1 and MTP loss. Additionally, PPY-PEI NZs induced lysosomal membrane permeabilization while inhibiting endosomal acidification, partly protecting cells from lysosomal apoptosis. PPY-PEI NZs selectively bound and eliminated exogenous malignant B cells in a mixed culture system with healthy leukocytes ex vivo. While PPY-PEI NZs showed no cytotoxicity in wild-type mice, they provided long-term and efficient inhibition of the growth of B-cell lymphoma-driven nodules in a subcutaneous xenograft model. This study explores a potential PPY-PEI NZ-based anticancer agent against B-cell lymphoma.
Collapse
|
40
|
Mahdieh A, Yeganeh H, Sande SA, Nyström B. Design of novel polyurethane-based ionene nanocarriers for cancer therapy: Synthesis, in-vitro, and in-vivo studies. Int J Pharm 2023; 635:122768. [PMID: 36841369 DOI: 10.1016/j.ijpharm.2023.122768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/01/2023] [Accepted: 02/19/2023] [Indexed: 02/26/2023]
Abstract
New strategies for constructing versatile nanocarriers are needed for cancer therapy to overcome the multiple challenges of targeted delivery. This work explores the advantages of polyurethane with main-chain quaternary ammonium salt moieties (ionene) as a novel carrier for targeted drug delivery. We have developed a novel cationic soybean oil-based polyurethane ionene nanocarrier (CPUI) that can act as an effective anticancer agent and efficiently deliver the anticancer drug 5-fluorouracil (5FU). We also report a potential anticancer drug delivery system targeting the folate receptor. In vitro experiments with blank CPUI carriers on the 4T1 (mouse breast cancer cell line) and the NIH-3T3 (mouse fibroblast cell line) revealed high cytotoxicity for the cancer cells but only low cytotoxicity for the normal fibroblast cells. The CPUI nanoparticles were readily loaded with 5FU (5FU-CPUI) in water using electrostatic interactions between the cationic quaternary ammonium groups of ionene and the anionic 5FU. The in vivo study in mice with tumors showed that the blank CPUI carriers significantly inhibited tumor growth, even more than the free drug (5FU). The inhibitory effect on tumor growth was slightly enhanced when the carriers were loaded with 5FU. The prepared nanoparticles had a high loading capacity of 41.8 %. Further enhancement of the inhibitory effect was observed when folic acid (FA) was added as a targeting moiety to the system via ion exchange with the bromine counterion of the quaternary ammonium moieties. The results suggest that the efficacy of FA-CPUI-5FU nanoparticles as vehicles for drug delivery can be enhanced via folate receptor (FR) mediated endocytosis in 4T1 cells and these novel nanocarriers may provide a potential platform for effective targeted drug delivery to tumor tissue and breast cancer therapy in the clinic.
Collapse
Affiliation(s)
- Athar Mahdieh
- Department of Pharmacy, Section for Pharmaceutics and Social Pharmacy, University of Oslo, Oslo, Norway
| | - Hamid Yeganeh
- Iran Polymer and Petrochemical Institute, Tehran, Iran.
| | - Sverre Arne Sande
- Department of Pharmacy, Section for Pharmaceutics and Social Pharmacy, University of Oslo, Oslo, Norway
| | - Bo Nyström
- Department of Chemistry, University of Oslo, Oslo, Norway.
| |
Collapse
|
41
|
Mahmoud SM, Ali SH, Omar MMA. Cationic cellulose nanocrystals as sustainable green material for multi biological applications via ξ potential. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2023:1-25. [PMID: 36752027 DOI: 10.1080/09205063.2023.2177474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The present study aims to disclose the activity of cationic cellulose nanocrystals (CNCs) as a promising multifunctional green nanomaterial with applications in biological aspects. The basic reason behind multifunctional behavior is zeta potential and size distribution of nano biopolymers; exhibit a remarkable physical and biological activity compared to normal molecules.The preliminary characterized studied using absorption spectral analysis showed strong absorption peak indicating that spectrum curves can be screen by UV spectra at wavelength range 200-400nm. Ultrastructural studies (SEM-EDS and TEM), manifest that CNCs are elliptical particles in shape. Also, TEM show CNCs are the ideal illustration of zero-dimensional (0-D) NPs, less than 5.1 nm in diameter with Cationic charge and similar results in size distribution by TEM. Nonetheless, developed as antioxidant activity IC50 was 1467 ± 25.9 µg/mL, antimicrobial activity tested G-ve strains, but not affected on tested G+ve strains and tested fungi. Evaluating toxicity effect of cationic CNCs against human blood erythrocytes (RBCs) and Lymphocyte Proliferation and the end point evaluate by comet assay, which proven no cytotoxic effect. Also, a high dose 500 µg/mL of CNCs highly significant (p < 0.05) reduction in cell viability of Caco-2 cancer cells after 24 h. incubation time, whereas the IC50 was 1884 ± 19.46 µg/mL. Moreover, genotoxic assay indicates Caco-2 cells cause apoptosis with no fragmentation in DNA. Undoubtedly, the obtained results brought about by the interaction of layers carrying opposing charges. Additionally, there is a balance between hydrophilic contact and electrostatic attraction. That emphasizes how the cationic CNCs have excellent potential for use as antioxidants, antimicrobials, and anticancer agents.
Collapse
Affiliation(s)
- Sara Mohamed Mahmoud
- Biotechnology Department, Faculty of Graduate Studies and Environmental Researches, Ain Shams University, Cairo, Egypt
| | - Safwat Hassan Ali
- Biochemistry Department, Faculty of Agriculture, Ain Shams University, Cairo, Egypt
| | - Mohamed M A Omar
- Biochemistry Department, Faculty of Agriculture, Ain Shams University, Cairo, Egypt
| |
Collapse
|
42
|
Manayia AH, Ilhami FB, Huang SY, Su TH, Huang CW, Chiu CW, Lee DJ, Lai JY, Cheng CC. Photoreactive Mercury-Containing Metallosupramolecular Nanoparticles with Tailorable Properties That Promote Enhanced Cellular Uptake for Effective Cancer Chemotherapy. Biomacromolecules 2023; 24:943-956. [PMID: 36645325 DOI: 10.1021/acs.biomac.2c01369] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
A new potential route to enhance the efficiency of supramolecular polymers for cancer chemotherapy was successfully demonstrated by employing a photosensitive metallosupramolecular polymer (Hg-BU-PPG) containing an oligomeric poly(propylene glycol) backbone and highly sensitive pH-responsive uracil-mercury-uracil (U-Hg-U) bridges. This route holds great promise as a multifunctional bioactive nano-object for development of more efficient and safer cancer chemotherapy. Owing to the formation of uracil photodimers induced by ultraviolet irradiation, Hg-BU-PPG can form a photo-cross-linked structure and spontaneously forms spherical nanoparticles in aqueous solution. The irradiated nanoparticles possess many unique characteristics, such as unique fluorescence behavior, highly sensitive pH-responsiveness, and intriguing phase transition behavior in aqueous solution as well as high structural stability and antihemolytic activity in biological media. More importantly, a series of cellular studies clearly confirmed that the U-Hg-U photo-cross-links in the irradiated nanoparticles substantially enhance their selective cellular uptake by cancer cells via macropinocytosis and the mercury-loaded nanoparticles subsequently induce higher levels of cytotoxicity in cancer cells (compared to non-irradiated nanoparticles), without harming normal cells. These results are mainly attributed to cancer cell microenvironment-triggered release of mercury ions from disassembled nanoparticles, which rapidly induce massive levels of apoptosis in cancer cells. Overall, the pH-sensitive U-Hg-U photo-cross-links within this newly discovered supramolecular system are an indispensable factor that offers a potential path to remarkably enhance the selective therapeutic effects of functional nanoparticles toward cancer cells.
Collapse
Affiliation(s)
- Abere Habtamu Manayia
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei10607, Taiwan
| | - Fasih Bintang Ilhami
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei10607, Taiwan.,Department of Natural Science, Faculty of Mathematics and Natural Science, Universitas Negeri Surabaya, Surabaya60231, Indonesia
| | - Sin-Yu Huang
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei10607, Taiwan
| | - Ting-Hsuan Su
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei10607, Taiwan
| | - Cheng-Wei Huang
- Department of Chemical and Materials Engineering, National Kaohsiung University of Science and Technology, Kaohsiung807618, Taiwan
| | - Chih-Wei Chiu
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei10607, Taiwan
| | - Duu-Jong Lee
- Department of Chemical Engineering, National Taiwan University, Taipei10617, Taiwan, Taiwan
| | - Juin-Yih Lai
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei10607, Taiwan.,Advanced Membrane Materials Research Center, National Taiwan University of Science and Technology, Taipei10607, Taiwan.,R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan32043, Taiwan.,Department of Chemical Engineering and Materials Science, Yuan Ze University, Chungli, Taoyuan32023, Taiwan
| | - Chih-Chia Cheng
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei10607, Taiwan.,Advanced Membrane Materials Research Center, National Taiwan University of Science and Technology, Taipei10607, Taiwan
| |
Collapse
|
43
|
Saccomandi P. Antimonene-Coated Uniform-Waist Tapered Fiber Optic Surface Plasmon Resonance Biosensor for the Detection of Cancerous Cells: Design and Optimization. ACS OMEGA 2023; 8:4627-4638. [PMID: 36777565 PMCID: PMC9909785 DOI: 10.1021/acsomega.2c06037] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Accepted: 12/05/2022] [Indexed: 06/18/2023]
Abstract
For early-stage cancer detection, a novel design of graphene-antimonene-coated uniform-waist tapered fiber optic surface plasmon resonance (SPR) biosensor is demonstrated. The proposed optical biosensor outperforms over a wide range of refractive index (RI) variations including biological solutions and is designed to detect various cancerous cells in the human body whose RIs are in the range of 1.36-1.4. Here, antimonene is used to enhance the performance of the designed SPR sensor for sensing cancer analytes because of its high binding energy toward adsorption of biomolecules and large active surface area. The design and analysis of the sensor are done with the help of a transfer matrix method-based simulation platform, and the effect of the taper ratio is also studied. The performance of the proposed SPR biosensor is evaluated with performance parameters such as sensitivity, full width at half maximum, detection accuracy (DA), figure of merit (FOM), and limit of detection (LOD). The numerical results show that the designed sensor is able to provide a sensitivity of 7.3465, 10.9250, 11.8914, and 15.2414 μm/RIU, respectively, for sensing skin, cervical, blood, and adrenal gland cancer with a maximum FOM of 131.1525 RIU-1, DA of 14.2126 μm-1, and LOD of 7.2 × 10-5 RIU. Based on the derived results, the authors believe that the designed SPR sensor could practically find its potential applications in the field of medical science for the early-stage diagnosis of cancer and hence, opens a new window in the field of biosensing.
Collapse
|
44
|
Cell Surface Charge Mapping Using a Microelectrode Array on ITO Substrate. Cells 2023; 12:cells12040518. [PMID: 36831185 PMCID: PMC9954061 DOI: 10.3390/cells12040518] [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: 12/16/2022] [Revised: 01/26/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Many cellular functions are regulated by cell surface charges, such as intercellular signaling and metabolism. Noninvasive measurement of surface charge distribution of a single cell plays a vital role in understanding cellular functions via cell membranes. We report a method for cell surface charge mapping via photoelectric interactions. A cell is placed on an array of microelectrodes fabricated on a transparent ITO (indium tin oxide) surface. An incident light irradiates the ITO surface from the backside. Because of the influence of the cell surface charge (or zeta potential), the photocurrent and the absorption of the incident light are changed, inducing a magnitude change of the reflected light. Hence, the cell surface charge distribution can be quantified by analyzing the reflected light intensity. This method does not need physical or chemical modification of the cell surface. We validated this method using charged microparticles (MPs) and two types of cells, i.e., human dermal fibroblast cells (HDFs) and human mesenchymal stem cells (hMSC). The measured average zeta potentials were in good agreement with the standard electrophoresis light scattering method.
Collapse
|
45
|
Donovan J, Deng Z, Bian F, Shukla S, Gomez-Arroyo J, Shi D, Kalinichenko VV, Kalin TV. Improving anti-tumor efficacy of low-dose Vincristine in rhabdomyosarcoma via the combination therapy with FOXM1 inhibitor RCM1. Front Oncol 2023; 13:1112859. [PMID: 36816948 PMCID: PMC9933126 DOI: 10.3389/fonc.2023.1112859] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023] Open
Abstract
Rhabdomyosarcoma (RMS) is a highly metastatic soft-tissue sarcoma that often develops resistance to current therapies, including vincristine. Since the existing treatments have not significantly improved survival, there is a critical need for new therapeutic approaches for RMS patients. FOXM1, a known oncogene, is highly expressed in RMS, and is associated with the worst prognosis in RMS patients. In the present study, we found that the combination treatment with specific FOXM1 inhibitor RCM1 and low doses of vincristine is more effective in increasing apoptosis and decreasing RMS cell proliferation in vitro compared to single drugs alone. Since RCM1 is highly hydrophobic, we developed innovative nanoparticle delivery system containing poly-beta-amino-esters and folic acid (NPFA), which efficiently delivers RCM1 to mouse RMS tumors in vivo. The combination of low doses of vincristine together with intravenous administration of NPFA nanoparticles containing RCM1 effectively reduced RMS tumor volumes, increased tumor cell death and decreased tumor cell proliferation in RMS tumors compared to RCM1 or vincristine alone. The combination therapy was non-toxic as demonstrated by liver metabolic panels using peripheral blood serum. Using RNA-seq of dissected RMS tumors, we identified Chac1 as a uniquely downregulated gene after the combination treatment. Knockdown of Chac1 in RMS cells in vitro recapitulated the effects of the combination therapy. Altogether, combination treatment with low doses of vincristine and nanoparticle delivery of FOXM1 inhibitor RCM1 in a pre-clinical model of RMS has superior anti-tumor effects and decreases CHAC1 while reducing vincristine toxicity.
Collapse
Affiliation(s)
- Johnny Donovan
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Zicheng Deng
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States,The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, United States,Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Fenghua Bian
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Samriddhi Shukla
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Jose Gomez-Arroyo
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States,Division of Pulmonary and Critical Care and Sleep Medicine, Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, United States
| | - Donglu Shi
- The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, United States
| | - Vladimir V. Kalinichenko
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States,Center for Lung Regenerative Medicine, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Tanya V. Kalin
- Division of Pulmonary Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States,*Correspondence: Tanya V. Kalin,
| |
Collapse
|
46
|
Nene LC, Nyokong T. The in-vitro proliferation-suppression of MCF-7 and HeLa cell lines mediated by differently substituted ionic phthalocyanines in sonodynamic therapy supplemented-photodynamic therapy. J Inorg Biochem 2023; 239:112084. [PMID: 36469974 DOI: 10.1016/j.jinorgbio.2022.112084] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/03/2022] [Accepted: 11/25/2022] [Indexed: 11/30/2022]
Abstract
This work focuses on the study of the effects of the ultrasonic frequency (MHz) and power (W.cm-2) on the stability, reactive oxygen species yields and cytotoxicity activities of differently substituted ionic phthalocyanines (Pcs) in sonodynamic therapy (SDT). Four ultrasonic parameters were investigated: Par I (1 MHz: 1 W.cm-2), Par II (1 MHz: 2 W.cm-2), Par III (3 MHz: 1 W.cm-2) and Par IV (3 MHz: 2 W.cm-2). A higher degradation of the Pcs was observed with increasing power at the Par II. Two reactive oxygen species (ROS) were detected in the ultrasound treated Pcs: singlet oxygen and hydroxyl radicals. Due to minimal degradation of most Pcs, Par I was chosen for SDT, photodynamic therapy (PDT), and photo-sonodynamic therapy (PSDT) against Michigan Cancer Foundation-7 and Henrietta Lacks cancer cell lines. PSDT generally showed improved therapeutic efficacies of the Pcs compared to the SDT and PDT mono treatments.
Collapse
Affiliation(s)
- Lindokuhle Cindy Nene
- Institute of Nanotechnology Innovation, P.O. 94, Rhodes University, Makhanda, South Africa
| | - Tebello Nyokong
- Institute of Nanotechnology Innovation, P.O. 94, Rhodes University, Makhanda, South Africa.
| |
Collapse
|
47
|
Enhancement of the in vitro anticancer photo-sonodynamic combination therapy activity of cationic thiazole-phthalocyanines using gold and silver nanoparticles. J Photochem Photobiol A Chem 2023. [DOI: 10.1016/j.jphotochem.2022.114339] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
48
|
Principle Superiority and Clinical Extensibility of 2D and 3D Charged Nanoprobe Detection Platform Based on Electrophysiological Characteristics of Circulating Tumor Cells. Cells 2023; 12:cells12020305. [PMID: 36672240 PMCID: PMC9856308 DOI: 10.3390/cells12020305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/21/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
The electrical characteristic of cancer cells is neglected among tumor biomarkers. The development of nanoprobes with opposing charges for monitoring the unique electrophysiological characteristics of cancer cells. Micro-nano size adsorption binding necessitates consideration of the nanoprobe's specific surface area. On the basis of the electrophysiological characteristics of circulating tumor cells (CTCs), clinical application and performance assessment are determined. To demonstrate that cancer cells have a unique pattern of electrophysiological patterns compared to normal cells, fluorescent nanoprobes with opposing charges were developed and fabricated. Graphene oxide (GO) was used to transform three-dimensional (3D) nanoprobes into two-dimensional (2D) nanoprobes. Compare 2D and 3D electrophysiological magnetic nanoprobes (MNP) in clinical samples and evaluate the adaptability and development of CTCs detection based on cell electrophysiology. Positively charged nanoprobes rapidly bind to negatively charged cancer cells based on electrostatic interactions. Compared to MNPs(+) without GO, the GO/MNPs(+) nanoprobe is more efficient and uses less material to trap cancer cells. CTCs can be distinguished from normal cells that are fully unaffected by nanoprobes by microscopic cytomorphological inspection, enabling the tracking of the number and pathological abnormalities of CTCs in the same patient at various chemotherapy phases to determine the efficacy of treatment. The platform for recognizing CTCs on the basis of electrophysiological characteristics compensates for the absence of epithelial biomarker capture and size difference capture in clinical performance. Under the influence of electrostatic attraction, the binding surface area continues to influence the targeting of cancer cells by nanoprobes. The specific recognition and detection of nanoprobes based on cell electrophysiological patterns has enormous potential in the clinical diagnosis and therapeutic monitoring of cancer.
Collapse
|
49
|
Tsoneva DK, Ivanov MN, Conev NV, Manev R, Stoyanov DS, Vinciguerra M. Circulating Histones to Detect and Monitor the Progression of Cancer. Int J Mol Sci 2023; 24:ijms24020942. [PMID: 36674455 PMCID: PMC9860657 DOI: 10.3390/ijms24020942] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 12/26/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Liquid biopsies have emerged as a minimally invasive cancer detection and monitoring method, which could identify cancer-related alterations in nucleosome or histone levels and modifications in blood, saliva, and urine. Histones, the core component of the nucleosome, are essential for chromatin compaction and gene expression modulation. Increasing evidence suggests that circulating histones and histone complexes, originating from cell death or immune cell activation, could act as promising biomarkers for cancer detection and management. In this review, we provide an overview of circulating histones as a powerful liquid biopsy approach and methods for their detection. We highlight current knowledge on circulating histones in hematologic malignancies and solid cancer, with a focus on their role in cancer dissemination, monitoring, and tumorigenesis. Last, we describe recently developed strategies to identify cancer tissue-of-origin in blood plasma based on nucleosome positioning, inferred from nucleosomal DNA fragmentation footprint, which is independent of the genetic landscape.
Collapse
Affiliation(s)
- Desislava K. Tsoneva
- Department of Medical Genetics, Faculty of Medicine, Medical University of Varna, 9000 Varna, Bulgaria
- Department of Stem Cell Biology and Transplantology, Research Institute, Medical University of Varna, 9000 Varna, Bulgaria
| | - Martin N. Ivanov
- Department of Stem Cell Biology and Transplantology, Research Institute, Medical University of Varna, 9000 Varna, Bulgaria
- Department of Anatomy and Cell Biology, Research Institute, Medical University of Varna, 9000 Varna, Bulgaria
| | - Nikolay Vladimirov Conev
- Clinic of Medical Oncology, UMHAT “St. Marina”, 1 “Hristo Smirnenski” Blvd., 9000 Varna, Bulgaria
- Department of Propedeutics of Internal Diseases, Medical University of Varna, 9000 Varna, Bulgaria
| | - Rostislav Manev
- Clinic of Medical Oncology, UMHAT “St. Marina”, 1 “Hristo Smirnenski” Blvd., 9000 Varna, Bulgaria
- Department of Propedeutics of Internal Diseases, Medical University of Varna, 9000 Varna, Bulgaria
| | - Dragomir Svetozarov Stoyanov
- Clinic of Medical Oncology, UMHAT “St. Marina”, 1 “Hristo Smirnenski” Blvd., 9000 Varna, Bulgaria
- Department of Propedeutics of Internal Diseases, Medical University of Varna, 9000 Varna, Bulgaria
| | - Manlio Vinciguerra
- Department of Stem Cell Biology and Transplantology, Research Institute, Medical University of Varna, 9000 Varna, Bulgaria
- Correspondence:
| |
Collapse
|
50
|
Naghaviyan A, Hashemi-Moghaddam H, Zavareh S, Ebrahimi Verkiani M, Meuller A. Synergistic Effect Evaluation of Magnetotherapy and a Cationic-Magnetic Nanocomposite Loaded with Doxorubicin for Targeted Drug Delivery to Breast Adenocarcinoma. Mol Pharm 2023; 20:101-117. [PMID: 36475680 DOI: 10.1021/acs.molpharmaceut.2c00505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This work investigates the synergistic effect of magnetotherapy and a novel cationic-magnetic drug delivery system on inhibiting breast cancer cell growth and other tissues. First, super-paramagnetic magnetite (Fe3O4) nanoparticles were coated with doxorubicin-imprinted poly(methacrylic acid-co-diallyl dimethylammonium chloride) [Fe3O4/poly(MAA-DDA)]. The cationic-magnetic nanocomposite (CMC) was characterized using XRD, FT-IR, VSM, TGA, TEM, FESEM, EDS, DLS, and BET. In vitro analyses, including drug release kinetics, cytotoxicity, and hemolytic assays, confirmed this novel CMC's good drug release profile and biocompatibility. Finally, in vivo experiments on BALB/c mice were designed to evaluate the synergistic effect of magnetotherapy on targeted drug delivery using the CMC. In vivo fluorescence imaging evaluated the drug distribution in different tissues of mice. Tumor volume evaluation demonstrated the efficiency of the CMC and magnetotherapy in preventing tumor growth; the two techniques significantly reduced tumor volume. Histopathological analysis proved that applying magnetotherapy in conjunction with the cationic-magnetic drug delivery system significantly prevented tumor cell proliferation and increased apoptosis with limited impact on other tissues. Also, Dox and Fe concentrations in different tissues confirmed the efficient drug delivery to tumor cells.
Collapse
Affiliation(s)
- Alireza Naghaviyan
- Department of Pharmacy, Damghan Branch, Islamic Azad University, 3671637849Damghan, Iran
| | | | - Saeed Zavareh
- School of Biology, Damghan University, 3671641167Damghan, Iran
| | | | - Anja Meuller
- Department of Chemistry and Biochemistry, Central Michigan University, Mount Pleasant, Michigan48859, United States
| |
Collapse
|