1
|
Tzou FY, Chuang PH, Hsu CH, Wu CH, Hsiao Y, Liu CC, Yu YL, Yeh YH, Lin CW, Chan CC, Huang SY. Dihydroceramide desaturase modulates autolysosome maturation and ameliorates CRB1 retinopathy. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167736. [PMID: 39965731 DOI: 10.1016/j.bbadis.2025.167736] [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: 09/02/2024] [Revised: 01/25/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
Variants in the CRB1 gene cause retinal degeneration and subsequent vision impairment in patients of retinitis pigmentosa (RP). No treatments are currently available to cure or impede the progression of CRB1-associated retinopathy. Previous studies have revealed alterations in the endolysosomal systems and autophagy in the absence of CRB1, but their roles in the pathogenesis of CRB1 retinopathy are unclear. Here, we examined the disease mechanism of CRB1 retinopathy using loss-of-function mutants of crumbs (crb), the Drosophila homolog of CRB1. We found that the loss of crb results in overactivation of autophagy in the eye. We also discovered that dihydroceramide desaturase encoded by infertile crescent (ifc), was up-regulated in crb mutants. Overexpression of ifc inhibited autolysosomes and alleviated Atg1-induced autophagic cell death. Mechanistically, ifc enhanced the binding of Rac1 to Atg8 and increased the autophagosomal localization of active Rac1, thus inhibiting autophagy. Importantly, autophagy inhibitions achieved through ifc overexpression, chloroquine treatment, or Beclin-1 RNAi all ameliorated the neurodegeneration of crb mutant eyes. Together, these findings highlight the mechanism of dihydroceramide desaturase in modulating autolysosome functions in crb mutants, providing new insights for developing treatments against CRB1 retinopathy.
Collapse
Affiliation(s)
- Fei-Yang Tzou
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Pei-Huan Chuang
- Department of Medical Research, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chia-Heng Hsu
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chih-Hsuan Wu
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yi Hsiao
- Department of Medical Research, National Taiwan University Hospital, Taipei 100, Taiwan
| | - Chung-Chih Liu
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yu-Lian Yu
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yu-Han Yeh
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chih-Wei Lin
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chih-Chaing Chan
- Graduate Institute of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Shu-Yi Huang
- Department of Medical Research, National Taiwan University Hospital, Taipei 100, Taiwan
| |
Collapse
|
2
|
Rawat J, Kachhadiya R, Thakkar H. Comparative in-vitro and in-vivo evaluation of spherulites and cubosomes of Irinotecan for lung targeting. Ther Deliv 2025; 16:339-347. [PMID: 39894804 PMCID: PMC11970735 DOI: 10.1080/20415990.2025.2460421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/27/2025] [Indexed: 02/04/2025] Open
Abstract
AIMS The present investigation aimed at the comparative evaluation of the developed nanocarriers, viz. spherulites and cubosomes for lung targeting. MATERIALS AND METHODS Both the spherulites and cubosomes were characterized for their entrapment efficiency, drug loading, size and zeta potential, in-vitro drug release profile, surface morphology, hemocompatibility, and in-vivo pharmacokinetic and lung biodistribution. RESULTS AND CONCLUSIONS The optimized batches of spherulites and cubosomes possessed high entrapment efficiency and drug loading with size around 200 nm, which is suitable for lung targeting. The zeta potential value for both the nanoformulations was found to be between -20 and -30 mv indicating the physical stability against aggregation. The SEM and TEM analysis revealed the presence of spherical and discrete particles in both the types of nanocarriers. Water channels were observed in case of cubosomes. Spherulites and cubosomes showed pH-dependent drug release with lower release at physiological pH while higher release at the pH of the tumor microenvironment. Both spherulites and cubosomes exhibited highly significant increase in the half-life and mean residence time in the plasma. The prepared nanoformulations were hemocompatible and had higher lung targeting potential compared to the plain drug solution.
Collapse
Affiliation(s)
- Jatin Rawat
- Centre for Relevance and Excellence in Novel drug delivery systems, Pharmacy Department, Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Fatehgunj, Vadodara, India
| | - Ravikumar Kachhadiya
- Centre for Relevance and Excellence in Novel drug delivery systems, Pharmacy Department, Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Fatehgunj, Vadodara, India
| | - Hetal Thakkar
- Centre for Relevance and Excellence in Novel drug delivery systems, Pharmacy Department, Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Fatehgunj, Vadodara, India
| |
Collapse
|
3
|
Malone K, LaCasse E, Beug ST. Cell death in glioblastoma and the central nervous system. Cell Oncol (Dordr) 2025; 48:313-349. [PMID: 39503973 PMCID: PMC11997006 DOI: 10.1007/s13402-024-01007-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2024] [Indexed: 04/15/2025] Open
Abstract
Glioblastoma is the commonest and deadliest primary brain tumor. Glioblastoma is characterized by significant intra- and inter-tumoral heterogeneity, resistance to treatment and dismal prognoses despite decades of research in understanding its biological underpinnings. Encompassed within this heterogeneity and therapy resistance are severely dysregulated programmed cell death pathways. Glioblastomas recapitulate many neurodevelopmental and neural injury responses; in addition, glioblastoma cells are composed of multiple different transformed versions of CNS cell types. To obtain a greater understanding of the features underlying cell death regulation in glioblastoma, it is important to understand the control of cell death within the healthy CNS during homeostatic and neurodegenerative conditions. Herein, we review apoptotic control within neural stem cells, astrocytes, oligodendrocytes and neurons and compare them to glioblastoma apoptotic control. Specific focus is paid to the Inhibitor of Apoptosis proteins, which play key roles in neuroinflammation, CNS cell survival and gliomagenesis. This review will help in understanding glioblastoma as a transformed version of a heterogeneous organ composed of multiple varied cell types performing different functions and possessing different means of apoptotic control. Further, this review will help in developing more glioblastoma-specific treatment approaches and will better inform treatments looking at more direct brain delivery of therapeutic agents.
Collapse
Affiliation(s)
- Kyle Malone
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Eric LaCasse
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada
| | - Shawn T Beug
- Apoptosis Research Centre, Children's Hospital of Eastern Ontario Research Institute, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada.
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
- Centre for Infection, Immunity and Inflammation, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
- Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Road, Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
4
|
Qiu Y, Wang Y, Liu J, Liu B, Sun K, Hou Q. Single-cell sequencing uncovers a high ESM1-expression endothelial cell subpopulation associated with bladder cancer progression and the immunosuppressive microenvironment. Sci Rep 2025; 15:10946. [PMID: 40159545 PMCID: PMC11955522 DOI: 10.1038/s41598-025-95731-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025] Open
Abstract
Despite remarkable advancements in therapeutic strategies, a considerable proportion of patients with bladder cancer (BC) still experience disease progression and unfavorable prognosis. The heterogeneity and biological functions of tumor endothelial cells (ECs) during BC progression remain poorly understood. We collected scRNA-seq data from BC samples and identified two EC subpopulations through hierarchical clustering analysis. The activity of signaling pathways in distinct EC subpopulations was assessed utilizing AUCell analysis. Gene regulatory networks (GRN) were constructed and analyzed for different EC subpopulations using the pySCENIC algorithm. Additionally, we investigated the association between the abundance of EC subpopulations and both clinical prognosis and immune cell infiltration. The biological effects of ESM1 protein on BC cells were further validated through EdU and Transwell assays. We analyzed 7,519 CD45-negative single cells from BC tissues and discerned two distinct EC subpopulations. The two subpopulations were characterized by high expression of ESM1 (S1 ECs) and CXCL2 (S2 ECs), respectively. In S1 ECs, we observed significant activation of signaling pathways involved in tumor promotion, including angiogenesis and cell proliferation. Additionally, our GRN analysis uncovered notable differences in transcription factor activity between S1 and S2 ECs. Moreover, ESM1 protein promoted proliferation and migration of BC cells. Patients with higher abundance of the S1 EC subpopulation exhibited more unfavorable clinical outcomes and increased infiltration of inhibitory immune cells. Our findings elucidate the transcriptional profiles and biological roles of the high ESM1-expression endothelial cell subpopulation in BC. This subpopulation is associated with poor prognosis and immunosuppressive tumor microenvironment. Accordingly, targeting endothelial cells with high ESM1 expression may offer a novel therapeutic strategy for patients with BC.
Collapse
Affiliation(s)
- Yifeng Qiu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
- International Cancer Center, Shenzhen Key Laboratory, Hematology Institution of ShenzhenUniversity, Shenzhen, China
| | - Yuhan Wang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Jiahe Liu
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical school, Shenzhen, 518060, China
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China
| | - Baohua Liu
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China.
| | - Kai Sun
- Department of Radiology, the Third People's Hospital of Longgang District, Shenzhen Clinical Medical School, Guangzhou University of Chinese Medicine, Shenzhen, 518116, China.
| | - Qi Hou
- Department of Urology, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, China.
- Shenzhen Key Laboratory for Systemic Aging and Intervention (SAI), Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, School of Basic Medical Sciences, Marshall Laboratory of Biomedical Engineering, National Engineering Research Center for Biotechnology (Shenzhen), International Cancer Center, Shenzhen University, Shenzhen, Guangdong, China.
- International Cancer Center, Shenzhen Key Laboratory, Hematology Institution of ShenzhenUniversity, Shenzhen, China.
| |
Collapse
|
5
|
Asghariazar V, Makaremi S, Amani N, Zare E, Kadkhodayi M, Eterafi M, Golmohammadi MG, Safarzadeh E. MicroRNA 320a-3p up-regulation reduces PD-L1 expression in gastric cancer cells: an experimental and bioinformatic study. Sci Rep 2025; 15:8239. [PMID: 40065071 PMCID: PMC11894147 DOI: 10.1038/s41598-025-92537-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Growing evidence suggests that dysregulated microRNAs were critical in the development of tumors and the progression number of malignancies. This research aimed to check the effect of microRNA 320a-3p transfection on gastric cancer (GC) cell lines. Following transfection, the efficacy was determined by the RT-PCR method. After that, MTT, scratch assay, DAPI staining, RT-PCR, and flow cytometry were used respectively. The results demonstrated that the viability of GC cells considerably decreased following transfection. Moreover, microRNA 320a-3p transfection significantly suppressed cell migration and induced apoptosis in these cells. We found that transfection of microRNA 320a-3p remarkably decreased PD-L1 gene expression and influenced epithelial-mesenchymal transition (EMT)-related and apoptotic gene expressions. The findings propose that microRNA 320a-3p could decrease cell proliferation and migration and induce apoptosis by increasing TP53 and CASP3 expression levels in GC cells. Notably, microRNA 320a-3p might be a potential target in GC immunotherapy by suppressing the PD-L1 gene expression.
Collapse
Affiliation(s)
- Vahid Asghariazar
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Deputy of Research and Technology, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shima Makaremi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Negin Amani
- School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Erfan Zare
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mahtab Kadkhodayi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Eterafi
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Mohammad Ghasem Golmohammadi
- Department of Anatomical Sciences and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
- Department of Microbiology, Parasitology, and Immunology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, 5166614711, Iran.
| |
Collapse
|
6
|
Ghosh R, Bhowmik A, Biswas S, Samanta P, Sarkar R, Pakhira S, Mondal M, Hajra S, Saha P. Natural flavonoid Orientin restricts 5-Fluorouracil induced cancer stem cells mediated angiogenesis by regulating HIF1α and VEGFA in colorectal cancer. Mol Med 2025; 31:85. [PMID: 40045186 PMCID: PMC11881437 DOI: 10.1186/s10020-024-01032-1] [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: 09/20/2024] [Accepted: 12/06/2024] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Cancer stem cells are a small subpopulation of cells which are responsible for tumor metastasis, angiogenesis, drug resistance etc. 5-Fluorouracil (5FU), a common therapeutic drug used in colorectal cancer treatment is reported to enrich CSCs, tumor recurrence and induces severe organ toxicities resulting in poor clinical outcome in patients. Therefore, we introduced a natural flavonoid Orientin in combination with 5FU to mitigate the CSC mediated angiogenesis and induced toxicities. METHODS Tumorosphere generation, flow cytometry, immunofluorescence assay, and western blotting were performed by using 5FU and Orientin individually and both treated colorectal cells and CSCs. In silico study was carried out to check the interaction between HIF1α and Orientin. In ovo chorioallantoic membrane (CAM) assay and tube formation assay using HUVECs were performed to monitor CSC mediated angiogenesis. In vivo CT26 syngeneic mice model was used to validate in silico and ex vivo results. RESULTS We found that 5FU treatment significantly increased the CD44+/CD133+ CSC population. In contrast, this CSC population in CSC enriched spheres (CES) derived from HCT116 cells were decreased by combination of Orientin and 5FU. Decrease of CSC's stemness properties was also noted, as evidenced by the downregulation of NANOG, SOX2 and OCT4. This new therapeutic strategy also inhibited CSC mediated angiogenesis by downregulating 5FU induced ROS, NO and LPO in those tumorospheres. Combination of Orientin and 5FU significantly reduced CSC mediated angiogenesis in HUVEC and CAM. Additionally, in silico study predicted that Orientin can bind to the PAS domain of HIF1α, a crucial factor for promoting angiogenesis. Expression of HIF1α and VEGFA were also decreased when the CESs were exposed to the combinatorial treatment. Additionally, we found that treatment with 5FU alone resulted reduction in tumor volume but it enriched CSCs and produced nephrotoxicity and hepatotoxicity in vivo. Combined treatment also considerably reduced the CD44+/CD133+ CSC population and hindered angiogenesis in a therapeutic in vivo model in BALB/c mice. CONCLUSIONS This novel treatment strategy of "Orientin with 5FU" is likely to improve the efficiency of conventional chemotherapy and may suppress disease recurrence in colorectal cancer by limiting CSC mediated angiogenesis.
Collapse
Affiliation(s)
- Rituparna Ghosh
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Arijit Bhowmik
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India.
| | - Souradeep Biswas
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Priya Samanta
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Rupali Sarkar
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Shampa Pakhira
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Mrinmoyee Mondal
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Subhadip Hajra
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Prosenjit Saha
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India.
| |
Collapse
|
7
|
Tesarova T, Fiala O, Hora M, Vaclavikova R. Non-coding transcriptome profiles in clear-cell renal cell carcinoma. Nat Rev Urol 2025; 22:151-174. [PMID: 39242964 DOI: 10.1038/s41585-024-00926-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/29/2024] [Indexed: 09/09/2024]
Abstract
Clear-cell renal cell carcinoma (ccRCC) is a common urological malignancy with an increasing incidence. The development of molecular biomarkers that can predict the response to treatment and guide personalized therapy selection would substantially improve patient outcomes. Dysregulation of non-coding RNA (ncRNA) has been shown to have a role in the pathogenesis of ccRCC. Thus, an increasing number of studies are being carried out with a focus on the identification of ncRNA biomarkers in ccRCC tissue samples and the connection of these markers with patients' prognosis, pathological stage and grade (including metastatic potential), and therapy outcome. RNA sequencing analysis led to the identification of several ncRNA biomarkers that are dysregulated in ccRCC and might have a role in ccRCC development. These ncRNAs have the potential to be prognostic and predictive biomarkers for ccRCC, with prospective applications in personalized treatment selection. Research on ncRNA biomarkers in ccRCC is advancing, but clinical implementation remains preliminary owing to challenges in validation, standardization and reproducibility. Comprehensive studies and integration of ncRNAs into clinical trials are essential to accelerate the clinical use of these biomarkers.
Collapse
Affiliation(s)
- Tereza Tesarova
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic.
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic.
| | - Ondrej Fiala
- Department of Oncology and Radiotherapeutics, Faculty of Medicine in Pilsen and University Hospital, Charles University, Pilsen, Czech Republic
- Laboratory of Cancer Treatment and Tissue Regeneration, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| | - Milan Hora
- Department of Urology, Faculty of Medicine in Pilsen and University Hospital, Charles University, Pilsen, Czech Republic
| | - Radka Vaclavikova
- Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic
| |
Collapse
|
8
|
Wakileh W, Watanabe NM, Amatsu Y, Sekiguchi H, Kajimura N, Kadonishi N, Umakoshi H. Investigation of Cubosome Interactions with Liposomal Membranes Based on Time-Resolved Small-Angle X-ray Scattering and Laurdan Fluorescence Spectroscopy. J Phys Chem B 2025. [PMID: 39999300 DOI: 10.1021/acs.jpcb.4c06060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2025]
Abstract
Nanosized dispersions of the bicontinuous cubic phase (cubosomes) are emerging carriers for drug delivery. These particles possess well-defined internal structures composed of highly-curved lipid bilayers that can accommodate significant drug payloads. Although cubosomes present promising potential for drug delivery, their physicochemical properties and interactions with cell membranes have not yet been fully understood. To clarify the interactions of the cubosomes with cell membranes, we investigated the changes in the structural and cubic membranes of monoolein (MO) cubosomes when mixed with model cell membranes at different phase states using time-resolved small-angle X-ray scattering (TR-SAXS), cryogenic transmission electron microscopy (cryo-TEM), and fluorescence spectroscopy. TR-SAXS results showed that the cubosomes gradually transitioned from the Im3m phase to the lamellar phase after interacting with the liposomes. The time of the structural change of the cubic phase to the lamellar phase was influenced by the fluidity of the liposome bilayers. Mixing the cubosomes with fluid membrane liposomes required less time to transition to the lamellar phase and vice versa. Cryo-TEM images showed that the well-defined internal structure of the cubosomes disappeared, leaving behind lamellar vesicles after the interaction, further confirming the TR-SAXS results. Laurdan fluorescence probe was used to assess the membrane polarity changes occurring to both the cubosomes and liposomes during the interaction. Examination of the normalized fluorescence intensity of the probed cubosomes showed decreasing intensity, followed by a recovery of intensity, which could indicate the disintegration of the cubic membrane and the formation of a mixed membrane. Also, the kinetics of the disintegration of the cubic phase did not seem to be influenced by the composition of the liposomes, which was in line with the normalized SAXS intensity results. Assessing the generalized polarization (GP340) values of the cubosomes and liposomes after mixing revealed that the fluidity and membrane hydration states of the cubosomes and liposomes transitioned to resemble their counterpart, confirming the exchange of material between the particles. Over time, the hydration states of the cubosomes and liposomes equilibrated toward an intermediate state between the two. The time needed to reach the final intermediate state was influenced by the membrane fluidity and hydration of the liposomes, more particularly the difference in GP340 values and their membrane phase state. These results highlight the importance of examination of the cubic membrane conditions, such as membrane polarity, and their implications on the changes in the cubic structure during the interaction with liposomal membranes.
Collapse
Affiliation(s)
- Ward Wakileh
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka 560-8531, Japan
| | - Nozomi Morishita Watanabe
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka 560-8531, Japan
| | - Yuki Amatsu
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka 560-8531, Japan
| | - Hiroshi Sekiguchi
- Japan Synchrotron Radiation Research Institute/SPring-8, Sayo, Hyogo 679-5198, Japan
| | - Naoko Kajimura
- Research Center for Ultra-High Voltage Electron Microscopy, Osaka University, 7-1, Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| | - Nanaki Kadonishi
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka 560-8531, Japan
| | - Hiroshi Umakoshi
- Division of Chemical Engineering, Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyamacho, Toyonaka, Osaka 560-8531, Japan
| |
Collapse
|
9
|
Ivashchenko O. Designing iron oxide & silver nanocomposites with phyto- and fungo chemicals for biomedicine: lessons learned. J Mater Chem B 2025; 13:1500-1517. [PMID: 39757969 DOI: 10.1039/d4tb02284j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Multifunctional nanoparticles for biomedical applications are widely researched and constantly developed because they provide wider possibilities for therapy and diagnostics. This work aims to summarise our findings towards the design of multifunctional complex iron oxide and silver nanoparticles (NPs) produced from the plants Zingiber officinale and Hypericum perforatum and mushrooms Amanita muscaria and Sparassis crispa. It was revealed that the antimicrobial and anticancer properties of the NPs were a consequence of the combination of silver and phyto- and fungo-chemicals originating from natural species. Moreover, the photoactive properties of the complex iron oxide and silver nanoparticles promoted photodynamic therapy (λexc = 405 nm) that significantly improved the antibacterial (E. coli, S. aureus, B. pumilus, P. fluorescence) and anticancer (HeLa, U2OS cells) effects. Notably, the gel formulations of the NPs based on hyaluronic and alginic acids had advantages over the aqueous dispersions of the NPs. For instance, being embedded into a hyaluronic acid gel, the NPs were more effective against cancer cells due to the improved uptake of hyaluronic acid by cancer cells. Another advantage of gel formulations of the NPs was connected with their microstructural properties; the nanocomposite gel adjusted its microstructure to the substrate topology, mimicking the substrate scale and pattern. Thus, complex ultrasmall iron oxide and silver nanoparticle NPs synthesized with natural extracts and their gel formulations may find diverse applications in the biomedical field, particularly for local cancer treatment and as post-operative bone or tissue scaffold after cancer or chronic osteomyelitis surgery.
Collapse
Affiliation(s)
- Olena Ivashchenko
- NanoBioMedical Centre, Adam Mickiewicz University in Poznań, 61614 Poznań, Poland.
| |
Collapse
|
10
|
Tüçer ME, Tunç N, Tüçer S, Acar R, Usta DD, Salimi K, Konu Ö, Şeker UÖŞ. Transcriptomic investigation of NP toxicity on HepaRG spheroids. Chem Biol Interact 2025; 405:111303. [PMID: 39515631 DOI: 10.1016/j.cbi.2024.111303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/11/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Metal nanoparticles (NPs) are commonly used nanomaterials, however concerns have been raised about their toxicity. Although a few studies have reported the toxicity of NPs on cells, they have generally been restricted to a limited variety of NPs, inappropriate cell lines, or culture methods. Thus, the adverse effects remain inadequately understood, necessitating further analysis. This study focuses on assessing the impacts of diverse NPs of varying materials and sizes on HepaRG spheroids to determine the genes that respond to acute NP toxicity. HepaRG cells, the most appropriate alternative to primary hepatocytes, were cultured in 3D spheroids to better mimic the cellular microenvironment of the liver. To elucidate the toxicity mechanisms of NPs on HepaRG spheroids, transcriptome analysis was conducted by using RNA sequencing (RNA-seq). Among all NPs, lowest and highest numbers of differentially expressed genes (DEGs) were found for 40 nm AuNP (118 genes) and InP/ZnS (1904 genes), respectively. Remarkably, processes such as drug metabolism, sensitivity to metal ions, oxidative stress, endothelial-mesenchymal transition (EMT) and apoptosis consistently exhibited significant enrichment across all NPs of different materials. Pathways related to stress responses of the cells such as the MAPK, p53 and mTOR pathways are found to be dysregulated upon exposure to various NPs. The genes that are common and unique between DEGs of different NPs were identified. These results provide novel insights into the toxicological mechanisms of NPs on HepaRG spheroids.
Collapse
Affiliation(s)
- Merve Erden Tüçer
- UNAM-Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey
| | - Nazlıcan Tunç
- UNAM-Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey
| | - Suat Tüçer
- UNAM-Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey
| | - Rana Acar
- Bilkent University, Department of Molecular Biology and Genetics, 06800, Ankara, Turkey
| | - Duygu Deniz Usta
- Gazi University, Faculty of Medicine, Department of Medical Biology and Genetics, 06500, Ankara, Turkey
| | - Kouroush Salimi
- Ankara Yildirim Beyazit University, Faculty of Engineering and Natural Sciences, Department of Chemical Engineering, 06010, Ankara, Turkey
| | - Özlen Konu
- UNAM-Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey; Bilkent University, Department of Molecular Biology and Genetics, 06800, Ankara, Turkey
| | - Urartu Özgür Şafak Şeker
- UNAM-Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey.
| |
Collapse
|
11
|
Louis D, Rizkalla CMZ, Rashad A. Cubosomes as Delivery System to Repositioning Nitrofurantoin in Breast Cancer Management. Drug Des Devel Ther 2024; 18:6173-6184. [PMID: 39722678 PMCID: PMC11668685 DOI: 10.2147/dddt.s499068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Purpose Nitrofurantoin (NITRO), a long-standing antibiotic to treat urinary tract infections, is activated by Nitro reductases. This activation mechanism has led to its exploration for repositioning applications in controlling and treating breast cancer, which express a Nitro reductase gene. Methods NITRO Cubosomes were developed using hot homogenization according to 23-full factorial design. The factors studied included the ratio of drug to oily phase (1:10 and 2:10), the ratio of oily to aqueous phase (1:10 and 1:5), and the ratio of Glyceryl mono-oleate (GMO) to Poloxamer 407 (PX407) (0.25:1 and 0.5:1). A total of 8 systems were proposed and evaluated by measuring particle size, zeta potential, polydispersity index, and percentage of entrapment efficiency. Results S6 (1:10 drug: oily phase, 1:5 oily: aqueous phase and 0.5:1 GMO: PX407) with particle size 45.5 ±c1.1 nm and an entrapment efficiency of 98.6 ± 1.8% exhibited highest desirability and was selected for further analysis. The morphology of S6 was examined using TEM microscopy. The activation of NITRO from S6 reflected on intracellular viability of MCF-7 breast cancer cell line was investigated by an MTT assay. The findings indicated that S6 had the lowest IC50 value (83.99 ± 0.15 μg g/mL) compared to Free NITRO (174.54 ± 1.36 μg g/mL), suggesting enhanced efficacy compared to free NITRO. Conclusion Nitrofurantoin cubosomes can be candidates for repositioning in breast cancer management after encouraging further stability and in-vivo studies.
Collapse
Affiliation(s)
- Dina Louis
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, Egypt
| | | | - Amira Rashad
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
| |
Collapse
|
12
|
Theodoropoulou MA, Mantzourani C, Kokotos G. Histone Deacetylase (HDAC) Inhibitors as a Novel Therapeutic Option Against Fibrotic and Inflammatory Diseases. Biomolecules 2024; 14:1605. [PMID: 39766311 PMCID: PMC11674560 DOI: 10.3390/biom14121605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/08/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Histone deacetylases (HDACs) are enzymes that play an essential role in the onset and progression of cancer. As a consequence, a variety of HDAC inhibitors (HDACis) have been developed as potent anticancer agents, several of which have been approved by the FDA for cancer treatment. However, recent accumulated research results have suggested that HDACs are also involved in several other pathophysiological conditions, such as fibrotic, inflammatory, neurodegenerative, and autoimmune diseases. Very recently, the HDAC inhibitor givinostat has been approved by the FDA for an indication beyond cancer: the treatment of Duchenne muscular dystrophy. In recent years, more and more HDACis have been developed as tools to understand the role that HDACs play in various disorders and as a novel therapeutic approach to fight various diseases other than cancer. In the present perspective article, we discuss the development and study of HDACis as anti-fibrotic and anti-inflammatory agents, covering the period from 2020-2024. We envision that the discovery of selective inhibitors targeting specific HDAC isozymes will allow the elucidation of the role of HDACs in various pathological processes and will lead to the development of promising treatments for such diseases.
Collapse
Affiliation(s)
- Maria A. Theodoropoulou
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (M.A.T.); (C.M.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Christiana Mantzourani
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (M.A.T.); (C.M.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece; (M.A.T.); (C.M.)
- Center of Excellence for Drug Design and Discovery, National and Kapodistrian University of Athens, 15771 Athens, Greece
| |
Collapse
|
13
|
Preman P, Moechars D, Fertan E, Wolfs L, Serneels L, Shah D, Lamote J, Poovathingal S, Snellinx A, Mancuso R, Balusu S, Klenerman D, Arranz AM, Fiers M, De Strooper B. APOE from astrocytes restores Alzheimer's Aβ-pathology and DAM-like responses in APOE deficient microglia. EMBO Mol Med 2024; 16:3113-3141. [PMID: 39528861 PMCID: PMC11628604 DOI: 10.1038/s44321-024-00162-7] [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: 04/24/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
The major genetic risk factor for Alzheimer's disease (AD), APOE4, accelerates beta-amyloid (Aβ) plaque formation, but whether this is caused by APOE expressed in microglia or astrocytes is debated. We express here the human APOE isoforms in astrocytes in an Apoe-deficient AD mouse model. This is not only sufficient to restore the amyloid plaque pathology but also induces the characteristic transcriptional pathological responses in Apoe-deficient microglia surrounding the plaques. We find that both APOE4 and the protective APOE2 from astrocytes increase fibrillar plaque deposition, but differentially affect soluble Aβ aggregates. Microglia and astrocytes show specific alterations in function of APOE genotype expressed in astrocytes. Our experiments indicate a central role of the astrocytes in APOE mediated amyloid plaque pathology and in the induction of associated microglia responses.
Collapse
Affiliation(s)
- Pranav Preman
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Daan Moechars
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Emre Fertan
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge, UK
| | - Leen Wolfs
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Lutgarde Serneels
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Disha Shah
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Jochen Lamote
- VIB FACS Expertise Center, Center for Cancer Biology, Leuven, Belgium
| | | | - An Snellinx
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - Renzo Mancuso
- Microglia and Inflammation in Neurological Disorders (MIND) Lab, VIB-UAntwerp, Centre for Molecular Neurology, Antwerp, Belgium
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Sriram Balusu
- VIB Center for Brain & Disease Research, Leuven, Belgium
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium
| | - David Klenerman
- Yusuf Hamied Department of Chemistry, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge, UK
| | - Amaia M Arranz
- Laboratory of Humanized Models of Disease, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, Spain
| | - Mark Fiers
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium.
| | - Bart De Strooper
- VIB Center for Brain & Disease Research, Leuven, Belgium.
- Laboratory for the Research of Neurodegenerative Diseases, Department of Neurosciences, Leuven Brain Institute (LBI), KU Leuven (University of Leuven), Leuven, Belgium.
- UK Dementia Research Institute, University College London, London, UK.
| |
Collapse
|
14
|
Abedimanesh S, Safaralizadeh R, Jahanafrooz Z, Najafi S, Amini M, Nazarloo SS, Bahojb Mahdavi SZ, Baradaran B, Jebelli A, Mokhtarzadeh AA. Interaction of noncoding RNAs with hippo signaling pathway in cancer cells and cancer stem cells. Noncoding RNA Res 2024; 9:1292-1307. [PMID: 39045083 PMCID: PMC11263728 DOI: 10.1016/j.ncrna.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 07/25/2024] Open
Abstract
The Hippo signaling pathway has a regulatory function in the organogenesis process and cellular homeostasis, switching the cascade reactions of crucial kinases acts to turn off/on the Hippo pathway, altering the downstream gene expression and thereby regulating proliferation, apoptosis, or stemness. Disruption of this pathway can lead to the occurrence of various disorders and different types of cancer. Recent findings highlight the importance of ncRNAs, such as microRNA, circular RNA, and lncRNAs, in modulating the Hippo pathway. Defects in ncRNAs can disrupt Hippo pathway balance, increasing tumor cells, tumorigenesis, and chemotherapeutic resistance. This review summarizes ncRNAs' inhibitory or stimulatory role in - Hippo pathway regulation in cancer and stem cells. Identifying the relation between ncRNAs and the components of this pathway could pave the way for developing new biomarkers in the treatment and diagnosis of cancers.
Collapse
Affiliation(s)
- Saba Abedimanesh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Zohreh Jahanafrooz
- Department of Biology, Faculty of Sciences, University of Maragheh, Maragheh, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shabnam Soltani Nazarloo
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asiyeh Jebelli
- Department of Biological Sciences, Faculty of Basic Sciences, Higher Education Institute of Rab-Rashid, Tabriz, Iran
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
15
|
Weiss FD, Alvarez Y, Shakeri F, Sahu A, Leka P, Dernst A, Rollheiser J, Vasconcelos M, Geraci A, Duthie F, Stahl R, Lee HE, Gellner AK, Buness A, Latz E, Meissner F. Retention of ES cell-derived 129S genome drives NLRP1 hypersensitivity and transcriptional deregulation in Nlrp3 tm1Flv mice. Cell Death Differ 2024; 31:1717-1729. [PMID: 39289506 PMCID: PMC11618613 DOI: 10.1038/s41418-024-01379-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024] Open
Abstract
Immune response genes are highly polymorphic in humans and mice, with heterogeneity amongst loci driving strain-specific host defence responses. The inadvertent retention of polymorphic loci can introduce confounding phenotypes, leading to erroneous conclusions, and impeding scientific advancement. In this study, we employ a combination of RNAseq and variant calling analyses to identify a substantial region of 129S genome, including the highly polymorphic Nlrp1 locus, proximal to Nlrp3, in one of the most commonly used mouse models of NLRP3 deficiency (Nlrp3tm1Flv). We show that the presence of the Nlrp1129S locus leads to an increase in NLRP1B protein expression, and a sensitising of Nlrp3tm1Flv macrophages to NLRP1 inflammasome activation, independent of NLRP3 deficiency. Retention of 129S genome further leads to protein sequence differences and altered gene regulation across multiple cell types, including of the key tissue-resident macrophage marker, TIM4. Using alternative models of NLRP3 deficiency, including a previously undescribed conditional Nlrp3 allele enabling precise temporal and cell-type specific control over Nlrp3 deletion, we further show that NLRP3 contributes to Talabostat-driven IL-1β release. Our study also establishes a generic framework to identify functionally relevant SNPs and assess genomic contamination in transgenic mice using RNAseq data. This allows for unambiguous attribution of phenotypes to the target gene and advances the precision and reliability of research in the field of host defence responses.
Collapse
Affiliation(s)
- Felix D Weiss
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany.
| | - Yubell Alvarez
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Farhad Shakeri
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University Hospital Bonn, Bonn, Germany
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Anshupa Sahu
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University Hospital Bonn, Bonn, Germany
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Petro Leka
- Institute of Innate Immunity, Department for Innate Immunity & Metaflammation, Medical Faculty, University of Bonn, Bonn, Germany
| | - Alesja Dernst
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Jessika Rollheiser
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Matilde Vasconcelos
- Institute of Innate Immunity, Department for Innate Immunity & Metaflammation, Medical Faculty, University of Bonn, Bonn, Germany
| | - Adriana Geraci
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Fraser Duthie
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Innate Immunity, Department for Innate Immunity & Metaflammation, Medical Faculty, University of Bonn, Bonn, Germany
| | - Rainer Stahl
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany
- Institute of Innate Immunity, Department for Innate Immunity & Metaflammation, Medical Faculty, University of Bonn, Bonn, Germany
| | - Hye Eun Lee
- Institute of Innate Immunity, Department for Innate Immunity & Metaflammation, Medical Faculty, University of Bonn, Bonn, Germany
| | - Anne-Kathrin Gellner
- Department of Psychiatry and Psychotherapy, University Hospital Bonn, Bonn, Germany
- Institute of Physiology II, Medical Faculty, University of Bonn, Bonn, Germany
| | - Andreas Buness
- Institute for Medical Biometry, Informatics and Epidemiology, Medical Faculty, University Hospital Bonn, Bonn, Germany
- Institute for Genomic Statistics and Bioinformatics, Medical Faculty, University of Bonn, Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, Department for Innate Immunity & Metaflammation, Medical Faculty, University of Bonn, Bonn, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
| | - Felix Meissner
- Institute of Innate Immunity, Department for Systems Immunology and Proteomics, Medical Faculty, University of Bonn, Bonn, Germany.
| |
Collapse
|
16
|
Wu H, Liu Y, Liu C. The interregulatory circuit between non-coding RNA and apoptotic signaling in diabetic cardiomyopathy. Noncoding RNA Res 2024; 9:1080-1097. [PMID: 39022683 PMCID: PMC11254508 DOI: 10.1016/j.ncrna.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 07/20/2024] Open
Abstract
Diabetes mellitus has surged in prevalence, emerging as a prominent epidemic and assuming a foremost position among prevalent medical disorders. Diabetes constitutes a pivotal risk element for cardiovascular maladies, with diabetic cardiomyopathy (DCM) standing out as a substantial complication encountered by individuals with diabetes. Apoptosis represents a physiological phenomenon observed throughout the aging and developmental stages, giving rise to the programmed cell death, which is implicated in DCM. Non-coding RNAs assume significant functions in modulation of gene expression. Their deviant expression of ncRNAs is implicated in overseeing diverse cellular attributes such as proliferation, apoptosis, and has been postulated to play a role in the progression of DCM. Notably, ncRNAs and the process of apoptosis can mutually influence and cooperate in shaping the destiny of human cardiac tissues. Therefore, the exploration of the interplay between apoptosis and non-coding RNAs holds paramount importance in the formulation of efficacious therapeutic and preventive approaches for managing DCM. In this review, we provide a comprehensive overview of the apoptotic signaling pathways relevant to DCM and subsequently delve into the reciprocal regulation between apoptosis and ncRNAs in DCM. These insights contribute to an enhanced comprehension of DCM and the development of therapeutic strategies.
Collapse
Affiliation(s)
- Hao Wu
- Public Health Clinical Center Affiliated to Shandong University, Jinan, 250100, China
| | - Yan Liu
- Public Health Clinical Center Affiliated to Shandong University, Jinan, 250100, China
| | - Chunli Liu
- Public Health Clinical Center Affiliated to Shandong University, Jinan, 250100, China
| |
Collapse
|
17
|
Eftekhari Kenzerki M, Mohajeri Khorasani A, Zare I, Amirmahani F, Ghasemi Y, Hamblin MR, Mousavi P. Deciphering the role of LOC124905135-related non-coding RNA cluster in human cancers: A comprehensive review. Heliyon 2024; 10:e39931. [PMID: 39641053 PMCID: PMC11617737 DOI: 10.1016/j.heliyon.2024.e39931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024] Open
Abstract
Non-coding RNAs (ncRNAs), especially microRNAs (miRNAs) and long ncRNAs (lncRNAs), are essential regulators of processes, such as the cell cycle and apoptosis. In addition to interacting with intracellular complexes and participating in diverse molecular pathways, ncRNAs can be used as clinical diagnostic biomarkers and therapeutic targets for fighting cancer. Studying ncRNA gene clusters is crucial for understanding their role in cancer and developing new treatments. LOC124905135 is a protein-coding gene encoding a collagen alpha-1(III) chain-like protein, and also acts as a gene for several ncRNAs, including miR-3619, PRR34 antisense RNA 1 (PRR34-AS1), PRR34, long intergenic ncRNA 2939 (LINC02939), LOC112268288, and MIRLET7BHG. It also serves as a host gene for three miRNAs (hsa-let7-A3, hsa-miR-4763, and hsa-let-7b). Notably, the ncRNAs derived from this particular genomic region significantly affect various cell functions, including the cell cycle and apoptosis. This cluster of ncRNAs is dysregulated in several types of cancer, exhibiting mutations, alterations in copy number, and being subject to DNA methylation and histone modification. In summary, the ncRNAs derived from the LOC124905135 cluster could be used as targets for diagnosis, therapy monitoring, and drug discovery in human cancers.
Collapse
Affiliation(s)
- Maryam Eftekhari Kenzerki
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Mohajeri Khorasani
- Department of Medical Genetics, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co., Ltd., Shiraz, 7178795844, Iran
| | - Farzane Amirmahani
- Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | - Younes Ghasemi
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy and Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Pegah Mousavi
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| |
Collapse
|
18
|
Zhou J, Guo M, Yang G, Cui X, Hu J, Lin T, Wang H, Gao S, Jiang C, Wang L, Wang Y. Chromatin landscape dynamics during reprogramming towards human naïve and primed pluripotency reveals the divergent function of PRDM1 isoforms. Cell Death Discov 2024; 10:474. [PMID: 39562537 PMCID: PMC11576854 DOI: 10.1038/s41420-024-02230-w] [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: 06/03/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/21/2024] Open
Abstract
Induced pluripotent stem cells (iPSCs) technology holds great potential in both scientific research and clinical applications. It enables the generation of naïve and primed iPSCs from various cell types through different strategies. Despite extensive characterizations of transcriptional and epigenetic factors, the intricacies of chromatin landscape dynamics during naïve and primed reprogramming, particularly in humans, remain poorly understood. In this study, we employed ATAC-seq and RNA-seq analyses to delineate and compare the chromatin landscape of naïve and primed pluripotency through the human secondary reprogramming system. Our investigations revealed several key transcriptional and epigenetic factors pivotal for reprogramming-associated chromatin remodeling. Notably, we found two isoforms of PRDM1, PRDM1α, and PRDM1β, bind to distinct genomic loci and play different roles in the naïve reprogramming process. We proposed an auto-regulatory model explaining the distinct functions of PRDM1α and PRDM1β. Overall, our findings highlight the complexity and diversity of transcription factors in shaping chromatin landscape dynamics and directing the fates of pluripotent cells.
Collapse
Affiliation(s)
- Jianfeng Zhou
- Shanghai Key Laboratory of Maternal and Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, 200092, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, 200092, Shanghai, China
| | - Mingyue Guo
- Frontier Science Center for Stem Cell Research, Tongji University, 200092, Shanghai, China
- The Fifth Affiliated Hospital of Guangzhou Medical University, 510700, Guangzhou, China
| | - Guang Yang
- Frontier Science Center for Stem Cell Research, Tongji University, 200092, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, 200065, Shanghai, China
| | - Xinyu Cui
- Frontier Science Center for Stem Cell Research, Tongji University, 200092, Shanghai, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, 200065, Shanghai, China
| | - Jindian Hu
- Shanghai Key Laboratory of Maternal and Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, 200092, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, 200092, Shanghai, China
| | - Tan Lin
- Shanghai Key Laboratory of Maternal and Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, 200092, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, 200092, Shanghai, China
| | - Hong Wang
- Shanghai Key Laboratory of Maternal and Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, 200092, Shanghai, China
- Frontier Science Center for Stem Cell Research, Tongji University, 200092, Shanghai, China
| | - Shaorong Gao
- Shanghai Key Laboratory of Maternal and Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, 200092, Shanghai, China.
- Frontier Science Center for Stem Cell Research, Tongji University, 200092, Shanghai, China.
| | - Cizhong Jiang
- Frontier Science Center for Stem Cell Research, Tongji University, 200092, Shanghai, China.
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of the Ministry of Education, Orthopaedic Department of Tongji Hospital, Tongji University, 200065, Shanghai, China.
| | - Liping Wang
- Frontier Science Center for Stem Cell Research, Tongji University, 200092, Shanghai, China.
- Shanghai Tenth People's Hospital, School of Life Sciences and Technology, Tongji University, 200072, Shanghai, China.
| | - Yixuan Wang
- Shanghai Key Laboratory of Maternal and Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, 200092, Shanghai, China.
- Frontier Science Center for Stem Cell Research, Tongji University, 200092, Shanghai, China.
| |
Collapse
|
19
|
Deng K, Hua Y, Gao Y, Zheng H, Jiang Y, Wang Y, Gao C, Ren T, Zhu Y. Thermosensitive Hydrogel with Programmable, Self-Regulated HIF-1α Stabilizer Release for Myocardial Infarction Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2408013. [PMID: 39308185 DOI: 10.1002/advs.202408013] [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: 07/14/2024] [Revised: 08/27/2024] [Indexed: 11/22/2024]
Abstract
HIF-1α (hypoxia induced factor-1α), a vital protective signal against hypoxia, has a short lifetime after myocardial infarction (MI). Increasing HIF-1α stability by inhibiting its hydroxylation with prolyl hydroxylases inhibitors such as DPCA (1,4-dihydrophenonthrolin-4-one-3-carboxylic acid) presents positive results. However, the optimal inhibitor administration profile for MI treatment is still unexplored. Here, injectable, thermosensitive hydrogels with programmable DPCA release are designed and synthesized. Hydrogel degradation and slow DPCA release are coupled to form a feedback loop by attaching pendant DPCA to polymer backbone, which serve as additional crosslinking points through π-π and hydrophobic interactions. Pendant carboxyl groups are added to the copolymer to accelerate DPCA release. Burst release in the acute phase for myocardial protection and extended near zero-order release across the inflammatory and fibrotic phases with different rates are achieved. All DPCA-releasing hydrogels upregulate HIF-1α, decrease apoptosis, promote angiogenesis, and stimulate cardiomyocyte proliferation, leading to preserved cardiac function and ventricular geometry. Faster hydrogel degradation induced by faster DPCA release results in a HIF-1α expression eight times of healthy control and better therapeutic effect in MI treatment. This research demonstrates the value of precise regulation of HIF-1α expression in treating MI and other relevant diseases and provides an implantable device-based modulation strategy.
Collapse
Affiliation(s)
- Kaicheng Deng
- State Key Laboratory of Transvascular Implantation Devices, Department of Cardiology of The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yuyan Hua
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ying Gao
- State Key Laboratory of Transvascular Implantation Devices, Department of Cardiology of The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, China
| | - Houwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yangzi Jiang
- School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China, Hong Kong, 999077, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China, Hong Kong, 999077, China
| | - Yaping Wang
- State Key Laboratory of Transvascular Implantation Devices, Department of Cardiology of The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, China
| | - Changyou Gao
- State Key Laboratory of Transvascular Implantation Devices, Department of Cardiology of The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Tanchen Ren
- State Key Laboratory of Transvascular Implantation Devices, Department of Cardiology of The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Heart Regeneration and Repair Key Laboratory of Zhejiang Province, Hangzhou, 310009, China
| | - Yang Zhu
- State Key Laboratory of Transvascular Implantation Devices, Department of Cardiology of The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
- Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| |
Collapse
|
20
|
Jang J, Jung H, Jeong J, Jeon J, Lee K, Jang HR, Han JW, Lee J. Modeling doxorubicin-induced-cardiotoxicity through breast cancer patient specific iPSC-derived heart organoid. Heliyon 2024; 10:e38714. [PMID: 39640743 PMCID: PMC11620051 DOI: 10.1016/j.heliyon.2024.e38714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 12/07/2024] Open
Abstract
Heart organoid (HO) technology has successfully overcome the limitations of two-dimensional (2D) disease modeling and drug testing, thereby emerging as a valuable tool in drug discovery for assessing toxicity and efficacy. However, its ability to distinguish drug responses among individuals remain unclear, which is crucial for developing predictive models. We addressed this gap by comparing human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) with human induced pluripotent stem cell-derived heart organoids (hiPSC-HOs) in the context of doxorubicin-induced cardiotoxicity (DIC). For this study, we utilized hiPSCs generated from breast cancer patients who had previously been treated with doxorubicin. By comparing groups with and without DIC, we examined various parameters, including cell viability, mRNA expression, protein expression and electrophysiological variations. The results of our analysis revealed significant differences between these groups, providing insights into hiPSC-HOs as a potential platform for testing differences in drug responses among patients.
Collapse
Affiliation(s)
- Jiye Jang
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyewon Jung
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jaekyun Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Junseok Jeon
- Division of Nephrology, Department of Medicine, Cell and Gene Therapy Institute, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Kyungho Lee
- Division of Nephrology, Department of Medicine, Cell and Gene Therapy Institute, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Hye Ryoun Jang
- Division of Nephrology, Department of Medicine, Cell and Gene Therapy Institute, Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - Jeung-Whan Han
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jaecheol Lee
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Epigenome Dynamics Control Research Center (EDCRC), School of Pharmacy, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Department of Biohealth Regulatory Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
21
|
Rahdan F, Saberi A, Saraygord-Afshari N, Hadizadeh M, Fayeghi T, Ghanbari E, Dianat-Moghadam H, Alizadeh E. Deciphering the multifaceted role of microRNAs in hepatocellular carcinoma: Integrating literature review and bioinformatics analysis for therapeutic insights. Heliyon 2024; 10:e39489. [PMID: 39498055 PMCID: PMC11532857 DOI: 10.1016/j.heliyon.2024.e39489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/06/2024] [Accepted: 10/15/2024] [Indexed: 11/07/2024] Open
Abstract
Hepatocellular carcinoma (HCC) poses a significant global health challenge, necessitating innovative therapeutic strategies. MicroRNAs (miRNAs) have emerged as pivotal regulators of HCC pathogenesis, influencing key processes such as self-renewal, angiogenesis, glycolysis, autophagy, and metastasis. This article integrates findings from a comprehensive literature review and bioinformatics analysis to elucidate the role of miRNAs in HCC. We discuss how dysregulation of miRNAs can drive HCC initiation, progression, and metastasis by modulating various signaling pathways and target genes. Moreover, leveraging high-throughput technology and bioinformatics tools, we identify key miRNAs involved in multiple cancer hallmarks, offering insights into potential combinatorial therapeutic strategies. Through our analysis considering p-values and signaling pathways associated with key features, we unveil miRNAs with simultaneous roles across critical cancer characteristics, providing a basis for the development of high-performance biomarkers. The microRNAs, miR-34a-5p, miR-373-3p, miR-21-5p, miR-214-5p, miR-195-5p, miR-139-5p were identified to be shared microRNAs in stemness, angiogenesis, glycolysis, autophagy, EMT, and metastasis of HCC. However, challenges such as miRNA stability and delivery hinder the translation of miRNA-based therapeutics into clinical practice. This review underscores the importance of further research to overcome existing barriers and realize the full potential of miRNA-based interventions for HCC management.
Collapse
Affiliation(s)
- Fereshteh Rahdan
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alihossein Saberi
- Department of Medical Genetics, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Neda Saraygord-Afshari
- Department of Medical Biotechnology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran, 1449614535, Iran
| | - Morteza Hadizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Tahura Fayeghi
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ghanbari
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, 8174673461, Iran
| | - Effat Alizadeh
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
22
|
Wang J, Zhan H, Wang Y, Zhao L, Huang Y, Wu R. Current advances in understanding endometrial epithelial cell biology and therapeutic applications for intrauterine adhesion. Stem Cell Res Ther 2024; 15:379. [PMID: 39456113 PMCID: PMC11515228 DOI: 10.1186/s13287-024-03989-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The human endometrium is a highly regenerative tissue capable of undergoing scarless repair during the menstruation and postpartum phases. This process is mediated by endometrial adult stem/progenitor cells. During the healing of endometrial injuries, swift reepithelization results in the rapid covering of the wound surface and facilitates subsequent endometrial restoration. The involvement of endogenous endometrial epithelial stem cells, stromal cells, and bone marrow-derived cells in the regeneration of the endometrial epithelium has been a subject of prolonged debate. Increasing evidence suggests that the regeneration of the endometrial epithelium mainly relies on epithelial stem cells rather than stromal cells and bone marrow-derived cells. Currently, no consensus has been established on the identity of epithelial stem cells in the epithelial compartment. Several markers, including stage-specific embryonic antigen-1 (SSEA-1), sex-determining region Y-box 9 (SOX9), neural-cadherin (N-cadherin), leucine-rich-repeat-containing G-protein-coupled receptor 5 (LGR5), CD44, axis inhibition protein 2 (Axin2), and aldehyde dehydrogenase 1A1 (ALDH1A1), have been suggested as potential candidate markers for endometrial epithelial stem cells. The identification of endometrial epithelial stem cells contributes to our understanding of endometrial regeneration and offers new therapeutic insights into diseases characterized by regenerative defects in the endometrium, such as intrauterine adhesion. This review explores different perspectives on the origins of human and mouse endometrial epithelial cells. It summarizes the potential markers, locations, and hierarchies of epithelial stem cells in both human and mouse endometrium. It also discusses epithelial cell-based treatments for intrauterine adhesion, hoping to inspire further research and clinical application of endometrial epithelial stem cells.
Collapse
Affiliation(s)
- Jia Wang
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, 310006, Zhejiang, People's Republic of China
- Zhejiang Key Laboratory of Maternal and Infant Health, Hangzhou, People's Republic of China
| | - Hong Zhan
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, 310006, Zhejiang, People's Republic of China
- Zhejiang Key Laboratory of Maternal and Infant Health, Hangzhou, People's Republic of China
| | - Yinfeng Wang
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, 310006, Zhejiang, People's Republic of China
- Zhejiang Key Laboratory of Maternal and Infant Health, Hangzhou, People's Republic of China
| | - Li Zhao
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, 310006, Zhejiang, People's Republic of China
- Zhejiang Key Laboratory of Maternal and Infant Health, Hangzhou, People's Republic of China
| | - Yunke Huang
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, 310006, Zhejiang, People's Republic of China
- Zhejiang Key Laboratory of Maternal and Infant Health, Hangzhou, People's Republic of China
| | - Ruijin Wu
- Women's Hospital, Zhejiang University School of Medicine, No. 1 Xueshi Road, Hangzhou, 310006, Zhejiang, People's Republic of China.
- Zhejiang Key Laboratory of Maternal and Infant Health, Hangzhou, People's Republic of China.
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, People's Republic of China.
| |
Collapse
|
23
|
Selvadoss A, Baby HM, Zhang H, Bajpayee AG. Harnessing exosomes for advanced osteoarthritis therapy. NANOSCALE 2024; 16:19174-19191. [PMID: 39323205 PMCID: PMC11799831 DOI: 10.1039/d4nr02792b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024]
Abstract
Exosomes are nanosized, lipid membrane vesicles secreted by cells, facilitating intercellular communication by transferring cargo from parent to recipient cells. This capability enables biological crosstalk across multiple tissues and cells. Extensive research has been conducted on their role in the pathogenesis of degenerative musculoskeletal diseases such as osteoarthritis (OA), a chronic and painful joint disease that particularly affects cartilage. Currently, no effective treatment exists for OA. Given that exosomes naturally modulate synovial joint inflammation and facilitate cartilage matrix synthesis, they are promising candidates as next generation nanocarriers for OA therapy. Recent advancements have focused on engineering exosomes through endogenous and exogenous approaches to enhance their joint retention, cartilage and chondrocyte targeting properties, and therapeutic content enrichment, further increasing their potential for OA drug delivery. Notably, charge-reversed exosomes that utilize electrostatic binding interactions with cartilage anionic aggrecan glycosaminoglycans have demonstrated the ability to penetrate the full thickness of early-stage arthritic cartilage tissue following intra-articular administration, maximizing their therapeutic potential. These exosomes offer a non-viral, naturally derived, cell-free carrier for OA drug and gene delivery applications. Efforts to standardize exosome harvest, engineering, and property characterization methods, along with scaling up production, will facilitate more efficient and rapid clinical translation. This article reviews the current state-of-the-art, explores opportunities for exosomes as OA therapeutics, and identifies potential challenges in their clinical translation.
Collapse
Affiliation(s)
- Andrew Selvadoss
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
| | - Helna M Baby
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Hengli Zhang
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| | - Ambika G Bajpayee
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA.
- Department of Bioengineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
24
|
Jahedi S, Wang L, Yorke JA, Watmough J. Finding Hopf bifurcation islands and identifying thresholds for success or failure in oncolytic viral therapy. Math Biosci 2024; 376:109275. [PMID: 39127095 DOI: 10.1016/j.mbs.2024.109275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/02/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
We model interactions between cancer cells and viruses during oncolytic viral therapy. One of our primary goals is to identify parameter regions that yield treatment failure or success. We show that the tumor size under therapy at a particular time is less than the size without therapy. Our analysis demonstrates two thresholds for the horizontal transmission rate: a "failure threshold" below which treatment fails, and a "success threshold" above which infection prevalence reaches 100% and the tumor shrinks to its smallest size. Moreover, we explain how changes in the virulence of the virus alter the success threshold and the minimum tumor size. Our study suggests that the optimal virulence of an oncolytic virus depends on the timescale of virus dynamics. We identify a threshold for the virulence of the virus and show how this threshold depends on the timescale of virus dynamics. Our results suggest that when the timescale of virus dynamics is fast, administering a more virulent virus leads to a greater reduction in the tumor size. Conversely, when the viral timescale is slow, higher virulence can induce oscillations with high amplitude in the tumor size. Furthermore, we introduce the concept of a "Hopf bifurcation Island" in the parameter space, an idea that has applications far beyond the results of this paper and is applicable to many mathematical models. We elucidate what a Hopf bifurcation Island is, and we prove that small Islands can imply very slowly growing oscillatory solutions.
Collapse
Affiliation(s)
- Sana Jahedi
- Department of Mathematics, University of Maryland, College Park, MD, United States; Institute for Physical Sciences and Technology, University of Maryland, College Park, MD, United States.
| | - Lin Wang
- Department of Mathematics and Statistics, University of New Brunswick, NB, Canada
| | - James A Yorke
- Department of Mathematics, University of Maryland, College Park, MD, United States; Institute for Physical Sciences and Technology, University of Maryland, College Park, MD, United States
| | - James Watmough
- Department of Mathematics and Statistics, University of New Brunswick, NB, Canada
| |
Collapse
|
25
|
Hsu CY, Faisal A, Jumaa SS, Gilmanova NS, Ubaid M, Athab AH, Mirzaei R, Karampoor S. Exploring the impact of circRNAs on cancer glycolysis: Insights into tumor progression and therapeutic strategies. Noncoding RNA Res 2024; 9:970-994. [PMID: 38770106 PMCID: PMC11103225 DOI: 10.1016/j.ncrna.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/18/2024] [Accepted: 05/04/2024] [Indexed: 05/22/2024] Open
Abstract
Cancer cells exhibit altered metabolic pathways, prominently featuring enhanced glycolytic activity to sustain their rapid growth and proliferation. Dysregulation of glycolysis is a well-established hallmark of cancer and contributes to tumor progression and resistance to therapy. Increased glycolysis supplies the energy necessary for increased proliferation and creates an acidic milieu, which in turn encourages tumor cells' infiltration, metastasis, and chemoresistance. Circular RNAs (circRNAs) have emerged as pivotal players in diverse biological processes, including cancer development and metabolic reprogramming. The interplay between circRNAs and glycolysis is explored, illuminating how circRNAs regulate key glycolysis-associated genes and enzymes, thereby influencing tumor metabolic profiles. In this overview, we highlight the mechanisms by which circRNAs regulate glycolytic enzymes and modulate glycolysis. In addition, we discuss the clinical implications of dysregulated circRNAs in cancer glycolysis, including their potential use as diagnostic and prognostic biomarkers. All in all, in this overview, we provide the most recent findings on how circRNAs operate at the molecular level to control glycolysis in various types of cancer, including hepatocellular carcinoma (HCC), prostate cancer (PCa), colorectal cancer (CRC), cervical cancer (CC), glioma, non-small cell lung cancer (NSCLC), breast cancer, and gastric cancer (GC). In conclusion, this review provides a comprehensive overview of the significance of circRNAs in cancer glycolysis, shedding light on their intricate roles in tumor development and presenting innovative therapeutic avenues.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan City, 71710, Taiwan
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, Arizona, 85004, USA
| | - Ahmed Faisal
- Department of Pharmacy, Al-Noor University College, Nineveh, Iraq
| | - Sally Salih Jumaa
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Nataliya Sergeevna Gilmanova
- Department of Prosthetic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Russia, Moscow
| | - Mohammed Ubaid
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Aya H. Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Rasoul Mirzaei
- Venom & Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal & Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Hama Faraj GS, Hussen BM, Abdullah SR, Fatih Rasul M, Hajiesmaeili Y, Baniahmad A, Taheri M. Advanced approaches of the use of circRNAs as a replacement for cancer therapy. Noncoding RNA Res 2024; 9:811-830. [PMID: 38590433 PMCID: PMC10999493 DOI: 10.1016/j.ncrna.2024.03.012] [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: 12/14/2023] [Revised: 03/18/2024] [Accepted: 03/29/2024] [Indexed: 04/10/2024] Open
Abstract
Cancer is a broad name for a group of diseases in which abnormal cells grow out of control and are characterized by their complexity and recurrence. Although there has been progress in cancer therapy with the entry of precision medicine and immunotherapy, cancer incidence rates have increased globally. Non-coding RNAs in the form of circular RNAs (circRNAs) play crucial roles in the pathogenesis, clinical diagnosis, and therapy of different diseases, including cancer. According to recent studies, circRNAs appear to serve as accurate indicators and therapeutic targets for cancer treatment. However, circRNAs are promising candidates for cutting-edge cancer therapy because of their distinctive circular structure, stability, and wide range of capabilities; many challenges persist that decrease the applications of circRNA-based cancer therapeutics. Here, we explore the roles of circRNAs as a replacement for cancer therapy, highlight the main challenges facing circRNA-based cancer therapies, and discuss the key strategies to overcome these challenges to improve advanced innovative therapies based on circRNAs with long-term health effects.
Collapse
Affiliation(s)
- Goran Sedeeq Hama Faraj
- Department of Medical Laboratory Science, Komar University of Science and Technology, Sulaymaniyah, 46001, Iraq
| | - Bashdar Mahmud Hussen
- Department of Biomedical Sciences, College of Science, Cihan University-Erbil, Erbil, Kurdistan Region, 44001, Iraq
- Department of Clinical Analysis, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, 44001, Iraq
| | - Snur Rasool Abdullah
- Medical Laboratory Science, Lebanese French University, Erbil, Kurdistan Region, 44001, Iraq
| | - Mohammed Fatih Rasul
- Department of Pharmaceutical Basic Science, Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | | | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Luo X, Gong HB, Li ZC, Li DD, Li ZX, Sun J, Yan CY, Huang RT, Feng Y, Chen SR, Cao YF, Liu M, Wang R, Huang F, Sun WY, Kurihara H, Duan WJ, Liang L, Jin W, Wu YP, He RR, Li YF. Phospholipid peroxidation in macrophage confers tumor resistance by suppressing phagocytic capability towards ferroptotic cells. Cell Death Differ 2024; 31:1184-1201. [PMID: 39103535 PMCID: PMC11369141 DOI: 10.1038/s41418-024-01351-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Ferroptosis holds significant potential for application in cancer therapy. However, ferroptosis inducers are not cell-specific and can cause phospholipid peroxidation in both tumor and non-tumor cells. This limitation greatly restricts the use of ferroptosis therapy as a safe and effective anticancer strategy. Our previous study demonstrated that macrophages can engulf ferroptotic cells through Toll-like receptor 2 (TLR2). Despite this advancement, the precise mechanism by which phospholipid peroxidation in macrophages affects their phagocytotic capability during treatment of tumors with ferroptotic agents is still unknown. Here, we utilized flow sorting combined with redox phospholipidomics to determine that phospholipid peroxidation in tumor microenvironment (TME) macrophages impaired the macrophages ability to eliminate ferroptotic tumor cells by phagocytosis, ultimately fostering tumor resistance to ferroptosis therapy. Mechanistically, the accumulation of phospholipid peroxidation in the macrophage endoplasmic reticulum (ER) repressed TLR2 trafficking to the plasma membrane and caused its retention in the ER by disrupting the interaction between TLR2 and its chaperone CNPY3. Subsequently, this ER-retained TLR2 recruited E3 ligase MARCH6 and initiated the proteasome-dependent degradation. Using redox phospholipidomics, we identified 1-steaoryl-2-15-HpETE-sn-glycero-3-phosphatidylethanolamine (SAPE-OOH) as the crucial mediator of these effects. Conclusively, our discovery elucidates a novel molecular mechanism underlying macrophage phospholipid peroxidation-induced tumor resistance to ferroptosis therapy and highlights the TLR2-MARCH6 axis as a potential therapeutic target for cancer therapy.
Collapse
Affiliation(s)
- Xiang Luo
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Hai-Biao Gong
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Zi-Chun Li
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Dong-Dong Li
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Zi-Xuan Li
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Jie Sun
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Chang-Yu Yan
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Rui-Ting Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Yue Feng
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou, 511443, China
| | - Shu-Rui Chen
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Yun-Feng Cao
- Shanghai Institute for Biomedical and Pharmaceutical Technologies, National Health Commission Key Laboratory of Reproduction Regulation, Shanghai, China
| | - Mingxian Liu
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou, 511443, China
| | - Rong Wang
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Feng Huang
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, China
| | - Wan-Yang Sun
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Hiroshi Kurihara
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Wen-Jun Duan
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Lei Liang
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Wen Jin
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Yan-Ping Wu
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China.
| | - Rong-Rong He
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China.
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China.
- School of Chinese Materia Medica and Yunnan Key Laboratory of Southern Medicinal Utilization, Yunnan University of Chinese Medicine, Kunming, 650500, China.
| | - Yi-Fang Li
- Guangdong Second Provincial General Hospital/Guangdong Engineering Research Center of Chinese Medicine & Disease Susceptibility/Guangzhou Key Laboratory of Traditional Chinese Medicine & Disease Susceptibility/Guangdong-Hong Kong-Macao Universities Joint Laboratory for the Internationalization of Traditional Chinese Medicine/International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE)/Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM and New Drugs Research/State Key Laboratory of Bioactive Molecules and Druggability Assessment/Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
28
|
Gawel AM, Betkowska A, Gajda E, Godlewska M, Gawel D. Current Non-Metal Nanoparticle-Based Therapeutic Approaches for Glioblastoma Treatment. Biomedicines 2024; 12:1822. [PMID: 39200286 PMCID: PMC11351974 DOI: 10.3390/biomedicines12081822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/24/2024] [Accepted: 08/09/2024] [Indexed: 09/02/2024] Open
Abstract
The increase in the variety of nano-based tools offers new possibilities to approach the therapy of poorly treatable tumors, which includes glioblastoma multiforme (GBM; a primary brain tumor). The available nanocomplexes exhibit great potential as vehicles for the targeted delivery of anti-GBM compounds, including chemotherapeutics, nucleic acids, and inhibitors. The main advantages of nanoparticles (NPs) include improved drug stability, increased penetration of the blood-brain barrier, and better precision of tumor targeting. Importantly, alongside their drug-delivery ability, NPs may also present theranostic properties, including applications for targeted imaging or photothermal therapy of malignant brain cells. The available NPs can be classified into two categories according to their core, which can be metal or non-metal based. Among non-metal NPs, the most studied in regard to GBM treatment are exosomes, liposomes, cubosomes, polymeric NPs, micelles, dendrimers, nanogels, carbon nanotubes, and silica- and selenium-based NPs. They are characterized by satisfactory stability and biocompatibility, limited toxicity, and high accumulation in the targeted tumor tissue. Moreover, they can be easily functionalized for the improved delivery of their cargo to GBM cells. Therefore, the non-metal NPs discussed here, offer a promising approach to improving the treatment outcomes of aggressive GBM tumors.
Collapse
Affiliation(s)
- Agata M. Gawel
- Department of Histology and Embryology, Medical University of Warsaw, Chalubinskiego 5, 02-004 Warsaw, Poland;
| | - Anna Betkowska
- Department of Cell Biology and Immunology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland; (A.B.); (E.G.); (M.G.)
| | - Ewa Gajda
- Department of Cell Biology and Immunology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland; (A.B.); (E.G.); (M.G.)
| | - Marlena Godlewska
- Department of Cell Biology and Immunology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland; (A.B.); (E.G.); (M.G.)
| | - Damian Gawel
- Department of Cell Biology and Immunology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland; (A.B.); (E.G.); (M.G.)
| |
Collapse
|
29
|
Czerwonka A, Kałafut J, Wang S, Anameric A, Przybyszewska-Podstawka A, Toriseva M, Nees M. The Notch inhibitor, FLI-06, increases the chemosensitivity of head and neck Squamous cell carcinoma cells to taxanes-based treatment. Biomed Pharmacother 2024; 177:116822. [PMID: 38906029 DOI: 10.1016/j.biopha.2024.116822] [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: 03/24/2024] [Revised: 05/21/2024] [Accepted: 05/26/2024] [Indexed: 06/23/2024] Open
Abstract
Aberration of Notch signaling is one of the key events involved in the development and progression of head and neck squamous cell carcinoma (HNSCC). The Notch pathway controls the tissue-specific differentiation of normal squamous epithelial cells and is frequently altered in squamous carcinomas, thus affecting their proliferation, growth, survival, and chemosensitivity or resistance against anti-cancer agents. In this study, we show that the use of novel, small-molecule inhibitors of Notch signaling, such as FLI-06, can have a beneficial effect on increasing the chemosensitivity of HNSCC to taxane-based chemotherapy. Inhibition of Notch signaling by FLI-06 alone virtually blocks the proliferation and growth of HNSCC cells in both 2D and 3D cultures and the zebrafish model, which is accompanied by down-regulation of key Notch target genes and proteins. Mechanistically, FLI-06 treatment causes cell cycle arrest in the G1-phase and induction of apoptosis in HNSCC, which is accompanied by increased c-JunS63 phosphorylation. Combining FLI-06 with Docetaxel shows a synergistic effect and partially blocks the cell growth of aggressive HNSCC cells via enhanced apoptosis and modification of c-JunS243 phosphorylation via GSK-3β inhibition. In conclusion, inhibition of Notch signaling in HNSCC cells that retain active Notch signaling significantly supports taxane-based anticancer activities via modulation of both the GSK-3β and the c-Jun.
Collapse
Affiliation(s)
- Arkadiusz Czerwonka
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin 20-093, Poland.
| | - Joanna Kałafut
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin 20-093, Poland
| | - Shaoxia Wang
- Institute of Biomedicine, Cancer Research Unit and FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Alinda Anameric
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin 20-093, Poland
| | | | - Mervi Toriseva
- Institute of Biomedicine, Cancer Research Unit and FICAN West Cancer Centre Laboratory, University of Turku and Turku University Hospital, Turku, Finland
| | - Matthias Nees
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, Lublin 20-093, Poland
| |
Collapse
|
30
|
Elvira-Blázquez D, Fernández-Justel JM, Arcas A, Statello L, Goñi E, González J, Ricci B, Zaccara S, Raimondi I, Huarte M. YTHDC1 m 6A-dependent and m 6A-independent functions converge to preserve the DNA damage response. EMBO J 2024; 43:3494-3522. [PMID: 38951610 PMCID: PMC11329685 DOI: 10.1038/s44318-024-00153-x] [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: 08/09/2023] [Revised: 05/07/2024] [Accepted: 06/05/2024] [Indexed: 07/03/2024] Open
Abstract
Cells have evolved a robust and highly regulated DNA damage response to preserve their genomic integrity. Although increasing evidence highlights the relevance of RNA regulation, our understanding of its impact on a fully efficient DNA damage response remains limited. Here, through a targeted CRISPR-knockout screen, we identify RNA-binding proteins and modifiers that participate in the p53 response. Among the top hits, we find the m6A reader YTHDC1 as a master regulator of p53 expression. YTHDC1 binds to the transcription start sites of TP53 and other genes involved in the DNA damage response, promoting their transcriptional elongation. YTHDC1 deficiency also causes the retention of introns and therefore aberrant protein production of key DNA damage factors. While YTHDC1-mediated intron retention requires m6A, TP53 transcriptional pause-release is promoted by YTHDC1 independently of m6A. Depletion of YTHDC1 causes genomic instability and aberrant cancer cell proliferation mediated by genes regulated by YTHDC1. Our results uncover YTHDC1 as an orchestrator of the DNA damage response through distinct mechanisms of co-transcriptional mRNA regulation.
Collapse
Affiliation(s)
- Daniel Elvira-Blázquez
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
| | - José Miguel Fernández-Justel
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
| | - Aida Arcas
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
- Clarivate, Barcelona, Spain
| | - Luisa Statello
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
| | - Enrique Goñi
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
| | - Jovanna González
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain
| | - Benedetta Ricci
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Sara Zaccara
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Ivan Raimondi
- New York Genome Center, New York, NY, USA.
- Weill Cornell Medicine, New York, NY, USA.
| | - Maite Huarte
- Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.
- Institute of Health Research of Navarra (IdiSNA), Pamplona, Spain.
| |
Collapse
|
31
|
Panwar A, Lye A, Musib D, Upadhyay A, Karankumar I, Devi PB, Pal M, Maity B, Roy M. Strategic design and development of a siderophore mimic: pioneering anticancer therapy via ROS generation and ferroptosis. Dalton Trans 2024; 53:12119-12127. [PMID: 38979715 DOI: 10.1039/d4dt01461h] [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: 07/10/2024]
Abstract
We designed a tris-catecholate-based siderophore mimic, H6-T-CATL, to selectively chelate iron(III) from mitochondrial cytochromes and other iron-containing proteins within cellular matrices. This strategic sequestration aims to trigger apoptosis or ferroptosis in cancer cells through the glutathione (GSH)-dependent release of reduced iron and subsequent ROS-mediated cytotoxicity. Synthesis of H6-T-CATL involved precise peptide coupling reactions. Using the Fe(III)-porphyrin model (Fe-TPP-Cl), akin to cytochrome c, we studied H6-T-CATL's ability to extract iron(III), yielding a binding constant (Krel) of 1014 for the resulting iron(III) complex (FeIII-T-CATL)3-. This complex readily underwent GSH-mediated reduction to release bioavailable iron(II), which catalyzed Fenton-like reactions generating hydroxyl radicals (˙OH), confirmed by spectroscopic analyses. Our research underscores the potential of H6-T-CATL to induce cancer cell death by depleting iron(III) from cellular metalloproteins, releasing pro-apoptotic iron(II). Evaluation across various cancer types, including normal cells, demonstrated H6-T-CATL's cytotoxicity through ROS production, mitochondrial dysfunction, and activation of ferroptosis and DNA damage pathways. These findings propose a novel mechanism for cancer therapy, leveraging endogenous iron stores within cells. H6-T-CATL emerges as a promising next-generation anticancer agent, exploiting iron metabolism vulnerabilities to induce selective cancer cell death through ferroptosis induction.
Collapse
Affiliation(s)
- Abhishek Panwar
- Department of Chemistry, National Institute of Technology Manipur, Langol, Imphal West, 795004, Manipur, India.
| | - Anushree Lye
- Department of Systems Biology, Center of Biomedical Research (CBMR), Raebareli Road, Lucknow 226014, Uttar Pradesh, India.
| | - Dulal Musib
- Department of Chemistry, National Institute of Technology Manipur, Langol, Imphal West, 795004, Manipur, India.
| | - Aarti Upadhyay
- Department of Inorganic and Physical Chemistry, Indian Institute of Science Bangalore, Bangalore-50012, Karnataka, India
| | - Irungbam Karankumar
- Department of Chemistry, National Institute of Technology Manipur, Langol, Imphal West, 795004, Manipur, India.
| | - Paonam Bebika Devi
- Department of Chemistry, National Institute of Technology Manipur, Langol, Imphal West, 795004, Manipur, India.
| | - Maynak Pal
- Department of Chemistry, National Institute of Technology Manipur, Langol, Imphal West, 795004, Manipur, India.
| | - Biswanath Maity
- Department of Systems Biology, Center of Biomedical Research (CBMR), Raebareli Road, Lucknow 226014, Uttar Pradesh, India.
- Department of Biological Sciences, Bose Institute Unified Academic Campus, EN80, Sector V, Bidhan Nagar, Kolkata - 700091, West Bengal, India
| | - Mithun Roy
- Department of Chemistry, National Institute of Technology Manipur, Langol, Imphal West, 795004, Manipur, India.
- Department of Chemistry, National Institute of Technology Agartala, Jirania, West Tripura, Agartala, 799046, India
| |
Collapse
|
32
|
Nógrádi B, Nógrádi-Halmi D, Erdélyi-Furka B, Kádár Z, Csont T, Gáspár R. Mechanism of motoneuronal and pyramidal cell death in amyotrophic lateral sclerosis and its potential therapeutic modulation. Cell Death Discov 2024; 10:291. [PMID: 38898006 PMCID: PMC11187107 DOI: 10.1038/s41420-024-02055-7] [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: 01/29/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder clinically characterized by muscle atrophy and progressive paralysis. Loss of motoneurons and pyramidal cells is thought to be the center piece of the complex and multifaceted ALS pathology, however, the exact mechanisms laying behind motoneuronal cell death in the spinal cord and motor cortex are still unknown. It was originally proposed that apoptosis plays a fundamental role in motoneuronal demise, nonetheless, later it became clear that other forms of regulated cell death, including necroptosis, pyroptosis, ferroptosis, and autophagy-dependent cell death, may also contribute to motoneuron loss. Over the past years, multiple studies aimed to improve our understanding of the contributory role of these mechanisms as well as to offer novel targets for potential therapeutic interventions. The pharmacological inhibition of the ferroptotic pathway and the modulation of the autophagic machinery seem to have particularly promising effects, reducing motoneuron loss and slowing disease progression in transgenic models of ALS. Nevertheless, the potential beneficial effects of necroptosis-targeting interventions were mostly disproven in the latest studies. In this review we aim to summarize the current view on regulated cell death mechanisms that lead to motoneuronal and pyramidal cell degeneration in ALS and showcase their applicability as future drug targets.
Collapse
Affiliation(s)
- Bernát Nógrádi
- Department of Neurology, Albert Szent-Györgyi Health Centre, University of Szeged, Szeged, Hungary
- Euan MacDonald Centre for Motor Neuron Disease Research, University of Edinburgh, Edinburgh, UK
| | - Dóra Nógrádi-Halmi
- Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Barbara Erdélyi-Furka
- Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Zalán Kádár
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Tamás Csont
- Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary
| | - Renáta Gáspár
- Department of Biochemistry, Albert Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary.
- Interdisciplinary Centre of Excellence, University of Szeged, Szeged, Hungary.
| |
Collapse
|
33
|
A Vahab S, Nair A, Raj D, G P A, P P S, S Kumar V. Cubosomes as versatile lipid nanocarriers for neurological disorder therapeutics: a comprehensive review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3729-3746. [PMID: 38095651 DOI: 10.1007/s00210-023-02879-7] [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: 07/01/2023] [Accepted: 11/29/2023] [Indexed: 05/23/2024]
Abstract
Cubosomes are novel vesicular drug delivery systems with lipidic liquid crystal nanoparticles formed of predetermined proportions of amphiphilic lipids. They have a honeycomb-like structure and are thermodynamically stable. These bicontinuous lipid layers are separated into two water-based channels internally that can be used by various bioactive substances, including drugs, proteins, and peptides. This complex structure is responsible for its high drug-loading capacity. Cubosomes are thought to be promising vehicles for various routes of administration because of their extraordinary characteristics, including bioadhesion, the capacity to encapsulate hydrophilic, and hydrophobic, as well as amphiphilic substances, high resistance to environmental stress, and their ability to achieve controlled release through modification. One of the essential elements for improving patient compliance is the ability of these well-defined nano-drug delivery systems to boost the effectiveness of targeting while lowering the side effects/toxicities of payloads. The large internal surface area, a sufficiently uncomplicated fabrication procedure, and biodegradability make it an attractive nano lipid carrier for drug delivery. This review outlines the recent advancement of cubosomes for managing various neurological disorders, highlighting their potential in this field.
Collapse
Affiliation(s)
- Safa A Vahab
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Ayushi Nair
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Devika Raj
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Akhil G P
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Sreelakshmi P P
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India
| | - Vrinda S Kumar
- Amrita School of Pharmacy, Amrita Institute of Medical Sciences & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, Kerala, 682041, India.
| |
Collapse
|
34
|
Hassanie H, Penteado AB, de Almeida LC, Calil RL, da Silva Emery F, Costa-Lotufo LV, Trossini GHG. SETDB1 as a cancer target: challenges and perspectives in drug design. RSC Med Chem 2024; 15:1424-1451. [PMID: 38799223 PMCID: PMC11113007 DOI: 10.1039/d3md00366c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 03/16/2024] [Indexed: 05/29/2024] Open
Abstract
Genome stability is governed by chromatin structural dynamics, which modify DNA accessibility under the influence of intra- and inter-nucleosomal contacts, histone post-translational modifications (PTMs) and variations, besides the activity of ATP-dependent chromatin remodelers. These are the main ways by which chromatin dynamics are regulated and connected to nuclear processes, which when dysregulated can frequently be associated with most malignancies. Recently, functional crosstalk between histone modifications and chromatin remodeling has emerged as a critical regulatory method of transcriptional regulation during cell destiny choice. Therefore, improving therapeutic outcomes for patients by focusing on epigenetic targets dysregulated in malignancies should help prevent cancer cells from developing resistance to anticancer treatments. For this reason, SET domain bifurcated histone lysine methyltransferase 1 (SETDB1) has gained a lot of attention recently as a cancer target. SETDB1 is a histone lysine methyltransferase that plays an important role in marking euchromatic and heterochromatic regions. Hence, it promotes the silencing of tumor suppressor genes and contributes to carcinogenesis. Some studies revealed that SETDB1 was overexpressed in various human cancer types, which enhanced tumor growth and metastasis. Thus, SETDB1 appears to be an attractive epigenetic target for new cancer treatments. In this review, we have discussed the effects of its overexpression on the progression of tumors and the development of inhibitor drugs that specifically target this enzyme.
Collapse
Affiliation(s)
- Haifa Hassanie
- School of Pharmaceutical Sciences, University of São Paulo Brazil
| | | | | | | | - Flávio da Silva Emery
- School of Pharmaceutical Sciences of the Ribeirão Preto, University of São Paulo Brazil
| | | | | |
Collapse
|
35
|
Hodgson L, Li Y, Iturria-Medina Y, Stratton JA, Wolf G, Krishnaswamy S, Bennett DA, Bzdok D. Supervised latent factor modeling isolates cell-type-specific transcriptomic modules that underlie Alzheimer's disease progression. Commun Biol 2024; 7:591. [PMID: 38760483 PMCID: PMC11101463 DOI: 10.1038/s42003-024-06273-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/01/2024] [Indexed: 05/19/2024] Open
Abstract
Late onset Alzheimer's disease (AD) is a progressive neurodegenerative disease, with brain changes beginning years before symptoms surface. AD is characterized by neuronal loss, the classic feature of the disease that underlies brain atrophy. However, GWAS reports and recent single-nucleus RNA sequencing (snRNA-seq) efforts have highlighted that glial cells, particularly microglia, claim a central role in AD pathophysiology. Here, we tailor pattern-learning algorithms to explore distinct gene programs by integrating the entire transcriptome, yielding distributed AD-predictive modules within the brain's major cell-types. We show that these learned modules are biologically meaningful through the identification of new and relevant enriched signaling cascades. The predictive nature of our modules, especially in microglia, allows us to infer each subject's progression along a disease pseudo-trajectory, confirmed by post-mortem pathological brain tissue markers. Additionally, we quantify the interplay between pairs of cell-type modules in the AD brain, and localized known AD risk genes to enriched module gene programs. Our collective findings advocate for a transition from cell-type-specificity to gene modules specificity to unlock the potential of unique gene programs, recasting the roles of recently reported genome-wide AD risk loci.
Collapse
Affiliation(s)
- Liam Hodgson
- School of Computer Science, McGill University, Montréal, QC, Canada
- Mila - Quebec Artificial Intelligence Institute, Montréal, QC, Canada
| | - Yue Li
- School of Computer Science, McGill University, Montréal, QC, Canada
| | - Yasser Iturria-Medina
- McConnell Brain Imaging Centre (BIC), MNI, Faculty of Medicine, McGill University, Montréal, Canada
- Neurology and Neurosurgery Department, Montreal Neurological Institute (MNI), Faculty of Medicine, McGill University, Montréal, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, McGill University, Montréal, Canada
| | - Jo Anne Stratton
- Neurology and Neurosurgery Department, Montreal Neurological Institute (MNI), Faculty of Medicine, McGill University, Montréal, Canada
| | - Guy Wolf
- Mila - Quebec Artificial Intelligence Institute, Montréal, QC, Canada
- Department of Mathematics & Statistics, Université de Montréal, Montréal, Canada
| | - Smita Krishnaswamy
- Department of Computer Science, Department of Genetics, Yale University, New Haven, CT, USA
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, USA
| | - Danilo Bzdok
- Mila - Quebec Artificial Intelligence Institute, Montréal, QC, Canada.
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montréal, QC, Canada.
- The Neuro - Montréal Neurological Institute, McConnell Brain Imaging Centre, Faculty of Medicine, McGill University, Montréal, QC, Canada.
| |
Collapse
|
36
|
Hussein R, Abou-Shanab AM, Badr E. A multi-omics approach for biomarker discovery in neuroblastoma: a network-based framework. NPJ Syst Biol Appl 2024; 10:52. [PMID: 38760476 PMCID: PMC11101461 DOI: 10.1038/s41540-024-00371-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 04/16/2024] [Indexed: 05/19/2024] Open
Abstract
Neuroblastoma (NB) is one of the leading causes of cancer-associated death in children. MYCN amplification is a prominent genetic marker for NB, and its targeting to halt NB progression is difficult to achieve. Therefore, an in-depth understanding of the molecular interactome of NB is needed to improve treatment outcomes. Analysis of NB multi-omics unravels valuable insight into the interplay between MYCN transcriptional and miRNA post-transcriptional modulation. Moreover, it aids in the identification of various miRNAs that participate in NB development and progression. This study proposes an integrated computational framework with three levels of high-throughput NB data (mRNA-seq, miRNA-seq, and methylation array). Similarity Network Fusion (SNF) and ranked SNF methods were utilized to identify essential genes and miRNAs. The specified genes included both miRNA-target genes and transcription factors (TFs). The interactions between TFs and miRNAs and between miRNAs and their target genes were retrieved where a regulatory network was developed. Finally, an interaction network-based analysis was performed to identify candidate biomarkers. The candidate biomarkers were further analyzed for their potential use in prognosis and diagnosis. The candidate biomarkers included three TFs and seven miRNAs. Four biomarkers have been previously studied and tested in NB, while the remaining identified biomarkers have known roles in other types of cancer. Although the specific molecular role is yet to be addressed, most identified biomarkers possess evidence of involvement in NB tumorigenesis. Analyzing cellular interactome to identify potential biomarkers is a promising approach that can contribute to optimizing efficient therapeutic regimens to target NB vulnerabilities.
Collapse
Affiliation(s)
- Rahma Hussein
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Ahmed M Abou-Shanab
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt
| | - Eman Badr
- Biomedical Sciences Program, University of Science and Technology, Zewail City of Science and Technology, Giza, 12578, Egypt.
- Faculty of Computers and Artificial Intelligence, Cairo University, Giza, 12613, Egypt.
| |
Collapse
|
37
|
Samy AM, Kandeil MA, Sabry D, Abdel-Ghany A, Mahmoud MO. From NAFLD to NASH: Understanding the spectrum of non-alcoholic liver diseases and their consequences. Heliyon 2024; 10:e30387. [PMID: 38737288 PMCID: PMC11088336 DOI: 10.1016/j.heliyon.2024.e30387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 04/04/2024] [Accepted: 04/25/2024] [Indexed: 05/14/2024] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become one of the most frequent chronic liver diseases worldwide in recent decades. Metabolic diseases like excessive blood glucose, central obesity, dyslipidemia, hypertension, and liver function abnormalities cause NAFLD. NAFLD significantly increases the likelihood of liver cancer, heart disease, and mortality, making it a leading cause of liver transplants. Non-alcoholic steatohepatitis (NASH) is a more advanced form of the disease that causes scarring and inflammation of the liver over time and can ultimately result in cirrhosis and hepatocellular carcinoma. In this review, we briefly discuss NAFLD's pathogenic mechanisms, their progression into NASH and afterward to NASH-related cirrhosis. It also covers disease epidemiology, metabolic mechanisms, glucose and lipid metabolism in the liver, macrophage dysfunction, bile acid toxicity, and liver stellate cell stimulation. Additionally, we consider the contribution of intestinal microbiota, genetics, epigenetics, and ecological factors to fibrosis progression and hepatocellular carcinoma risk in NAFLD and NASH patients.
Collapse
Affiliation(s)
- Ahmed M. Samy
- Department of Biochemistry, Faculty of Pharmacy, Nahda University, Beni-Suef 62513, Egypt
| | - Mohamed A. Kandeil
- Department of Biochemistry, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Dina Sabry
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Badr University in Cairo, Cairo 11829, Egypt
| | - A.A. Abdel-Ghany
- Department of Biochemistry, Faculty of Pharmacy, Nahda University, Beni-Suef 62513, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Assuit Branch, Egypt
| | - Mohamed O. Mahmoud
- Department of Biochemistry, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
38
|
Cao MY, Zainudin S, Daud KM. Protein features fusion using attributed network embedding for predicting protein-protein interaction. BMC Genomics 2024; 25:466. [PMID: 38741045 DOI: 10.1186/s12864-024-10361-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Protein-protein interactions (PPIs) hold significant importance in biology, with precise PPI prediction as a pivotal factor in comprehending cellular processes and facilitating drug design. However, experimental determination of PPIs is laborious, time-consuming, and often constrained by technical limitations. METHODS We introduce a new node representation method based on initial information fusion, called FFANE, which amalgamates PPI networks and protein sequence data to enhance the precision of PPIs' prediction. A Gaussian kernel similarity matrix is initially established by leveraging protein structural resemblances. Concurrently, protein sequence similarities are gauged using the Levenshtein distance, enabling the capture of diverse protein attributes. Subsequently, to construct an initial information matrix, these two feature matrices are merged by employing weighted fusion to achieve an organic amalgamation of structural and sequence details. To gain a more profound understanding of the amalgamated features, a Stacked Autoencoder (SAE) is employed for encoding learning, thereby yielding more representative feature representations. Ultimately, classification models are trained to predict PPIs by using the well-learned fusion feature. RESULTS When employing 5-fold cross-validation experiments on SVM, our proposed method achieved average accuracies of 94.28%, 97.69%, and 84.05% in terms of Saccharomyces cerevisiae, Homo sapiens, and Helicobacter pylori datasets, respectively. CONCLUSION Experimental findings across various authentic datasets validate the efficacy and superiority of this fusion feature representation approach, underscoring its potential value in bioinformatics.
Collapse
Affiliation(s)
- Mei-Yuan Cao
- Center for Artificial Intelligence Technology (CAIT), Faculty of Information Science and Technology, Universiti Kebangsaan Malaysia, Bangi, 43600, Selangor, Malaysia.
| | - Suhaila Zainudin
- Center for Artificial Intelligence Technology (CAIT), Faculty of Information Science and Technology, Universiti Kebangsaan Malaysia, Bangi, 43600, Selangor, Malaysia
| | - Kauthar Mohd Daud
- Center for Artificial Intelligence Technology (CAIT), Faculty of Information Science and Technology, Universiti Kebangsaan Malaysia, Bangi, 43600, Selangor, Malaysia
| |
Collapse
|
39
|
Jain H, Prabhakar B, Shende PK. Potentiation of Brain Bioavailability Using Thermoreversible Cubosomal Formulation. Mol Pharm 2024; 21:2534-2543. [PMID: 38547474 DOI: 10.1021/acs.molpharmaceut.4c00085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2024]
Abstract
The aim of the present study was to develop and evaluate intranasal formulations of the thermoreversible fluoxetine cubosomal in situ gel. This gel was intended for permeation and bioavailability enhancement to target the brain effectively by bypassing the blood-brain barrier (BBB). Fluoxetine-loaded cubosomes were prepared by the homogenization method followed by the cold method approach to develop in situ gel. Fluoxetine-loaded cubosomes displayed a higher encapsulation efficiency (82.60 ± 1.25%) than fluoxetine. This might be due to the solubilizing activity of the polymer to cause partitioning of the lipophilic drug into the aqueous phase during the change from the cubic gel phase to cubosomes. In vitro analysis of fluoxetine-loaded cubosomal in situ gel showed a sustained release profile (93.22 ± 2.47%) due to limited diffusion of fluoxetine. The formation of strong affinity bonds of the drug with GMO (drug transporter) decreased the drug release in comparison to that with fluoxetine-loaded cubosomes (90.68 ± 1.74%). The ex vivo drug release profile revealed the drug release of 96.31 ± 2.88% by the end of 24 h. This is attributed to the higher capability of the intranasal cubosomal in situ gel to prolong the retention and enable better permeation through the nasal mucosa. In male Wistar rats, in vivo biodistribution studies for cubosomal in situ gel administered via the intranasal route at a dose of 3.5 mg/kg demonstrated an increase in pharmacokinetic parameters like the AUC (406 ± 75.35 μg/mL), Cmax (368.07 ± 0.23 μg/mL), Tmax (4 h), and t1/2 (14.06 h). The mucoadhesive nature of the in situ gel led to an increase in the residence time of the gel in the nasal mucosa. The biodistribution study of intranasal in situ cubosomal gel improved the bioavailability 2.21-fold in comparison to that with the cubosomal dispersion but 2.83-fold in comparison to that with the drug solution. Therefore, fluoxetine-loaded cubosomal in situ gel proved as a promising carrier for effective transportation of fluoxetine via the intranasal route with significant brain bioavailability.
Collapse
Affiliation(s)
- Harshvardhan Jain
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| | - Bala Prabhakar
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| | - Pravin K Shende
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V. L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| |
Collapse
|
40
|
Zahiri M, Kamali H, Abnous K, Mohammad Taghdisi S, Nekooei S, Nekooei N, Ramezani M, Alibolandi M. Synthesis of folate targeted theranostic cubosomal platform for co-delivery of bismuth oxide and doxorubicin to melanoma in vitro and in vivo. Eur J Pharm Biopharm 2024; 198:114259. [PMID: 38479563 DOI: 10.1016/j.ejpb.2024.114259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/21/2024] [Accepted: 03/11/2024] [Indexed: 04/19/2024]
Abstract
Liquid crystalline nanoparticles (LCNPs) have gained much attention in cancer nanomedicines due to their unique features such as high surface area, storage stability, and sustained-release profile. In the current study, a novel LCNP for co-encapsulation of Bi2O3 and hydrophilic doxorubicin (DOX) was fabricated and functionalized with folic acid (FA) to achieve efficient tumor targeting toward CT-scan imaging and chemotherapy of melanoma in vitro and in vivo. LCNPs Bi2O3 NPs were prepared using glycerol monooleate-pluronic F-127 (GMO/PF127/water). Firstly, GMO/water were homogenized to prepare LC gel. Then, the stabilizer aqueous solution (PF127/Bi2O3/DOX) was added to the prepared LC gel and homogenized using homogenization and ultrasonication. The formulated NPs exhibited superior stability with encapsulation efficiency. High cytotoxicity and cellular internalization of the FA-Bi2O3-DOX-NPs were observed in comparison with Bi2O3-DOX-NPs and the free DOX in folate-receptor (FR) overexpressing cells (B16F10) in vitro. Moreover, ideal tumor suppression with increased survival rate were observed in tumorized mice treated with FA-Bi2O3-DOX-NPs compared to those treated with non-targeted one. On the other hand, the CT-imaging ability of the Bi2O3-DOX-NPs was tested inB16F10 tumor-bearing mice. The obtained data indicated a high potential of the developed targeted theranostic FA-Bi2O3-DOX-NPs for diagnostics and treatment of melanoma.
Collapse
Affiliation(s)
- Mahsa Zahiri
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hosein Kamali
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sirous Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
41
|
Jori C, Ansari MM, Ahmad A, Ali N, Raza SS, Khan R. Biomaterial-based combinatorial approach of aescin-comprised zein-coated gelatin nanoparticles alleviates synovial inflammation in experimental inflammatory arthritis. NANOSCALE 2024; 16:7965-7975. [PMID: 38567436 DOI: 10.1039/d3nr06476j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease that mostly affects joints. Although RA therapy has made significant progress, difficulties including extensive medication metabolism and its quick clearance result in its inadequate bioavailability. The anti-inflammatory effect of zein was reported with other medications, but it has certain limitations. There are reports on the anti-oxidant and anti-inflammatory effect of aescin, which exhibits low bioavailability for the treatment of rheumatoid arthritis. Also, the combinatorial effect of zein with other effective drug delivery systems is still under investigation for the treatment of experimental collagen-induced rheumatoid arthritis. The focus of this study was to formulate and define the characteristics of zein-coated gelatin nanoparticles encapsulated with aescin (Ze@Aes-GNPs) and to assess and contrast the therapeutic effectiveness of Ze@Aes-GNPs towards collagen-induced RA in Wistar rats. Nanoprecipitation and the layer-by-layer coating process were used to fabricate Ze@Aes-GNPs and their hydrodynamic diameter was determined to be 182 nm. Scanning electron microscopy (SEM) and transmission electron microscopy (TEM) were used to further validate the size, shape, and surface morphology of Ze@Aes-GNPs. When tested against foreskin fibroblasts (BJ), these nanoparticles demonstrated significantly high cytocompatibility. Both Aes and Ze@Aes-GNPs were effective in treating arthritis, as shown by the decreased edoema, erythema, and swelling of the joints, between which Ze@Aes-GNPs were more effective. Further, it was demonstrated that Aes and Ze@Aes-GNPs reduced the levels of oxidative stress (articular elastase, lipid peroxidation, catalase, superoxide dismutase and nitric oxide) and inflammatory indicators (TNF-α, IL-1β and myeloperoxidase). The histopathology findings further demonstrated that Ze@Aes-GNPs considerably reduced the infiltration of inflammatory cells at the ankle joint cartilage compared to Aes. Additionally, immunohistochemistry examination showed that treatment with Ze@Aes-GNPs suppressed the expression of pro-inflammatory markers (COX-2 and IL-6) while increasing the expression of SOD1. In summary, the experiments indicated that Aes and Ze@Aes-GNPs lowered the severity of arthritis, and critically, Ze@Aes-GNPs showed better effectiveness in comparison to Aes. This suppression of oxidative stress and inflammation was likely driven by Aes and Ze@Aes-GNPs.
Collapse
Affiliation(s)
- Chandrashekhar Jori
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India.
| | - Md Meraj Ansari
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India.
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Nemat Ali
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Syed Shadab Raza
- Laboratory for Stem Cell and Restorative Neurology, Department of Biotechnology, Era's Lucknow Medical College Hospital, Sarfarazganj, Lucknow 226003, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India.
| |
Collapse
|
42
|
Nica I, Volovat C, Boboc D, Popa O, Ochiuz L, Vasincu D, Ghizdovat V, Agop M, Volovat CC, Lupascu Ursulescu C, Lungulescu CV, Volovat SR. A Holographic-Type Model in the Description of Polymer-Drug Delivery Processes. Pharmaceuticals (Basel) 2024; 17:541. [PMID: 38675501 PMCID: PMC11053585 DOI: 10.3390/ph17040541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/13/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
A unitary model of drug release dynamics is proposed, assuming that the polymer-drug system can be assimilated into a multifractal mathematical object. Then, we made a description of drug release dynamics that implies, via Scale Relativity Theory, the functionality of continuous and undifferentiable curves (fractal or multifractal curves), possibly leading to holographic-like behaviors. At such a conjuncture, the Schrödinger and Madelung multifractal scenarios become compatible: in the Schrödinger multifractal scenario, various modes of drug release can be "mimicked" (via period doubling, damped oscillations, modulated and "chaotic" regimes), while the Madelung multifractal scenario involves multifractal diffusion laws (Fickian and non-Fickian diffusions). In conclusion, we propose a unitary model for describing release dynamics in polymer-drug systems. In the model proposed, the polymer-drug dynamics can be described by employing the Scale Relativity Theory in the monofractal case or also in the multifractal one.
Collapse
Affiliation(s)
- Irina Nica
- Department of Odontology-Periodontology, Fixed Prosthesis, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Constantin Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str, 700115 Iasi, Romania;
| | - Diana Boboc
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str, 700115 Iasi, Romania;
| | - Ovidiu Popa
- Department of Emergency Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Lacramioara Ochiuz
- Faculty of Pharmacy, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Decebal Vasincu
- Department of Biophysics, Faculty of Dental Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Vlad Ghizdovat
- Department of Biophysics and Medical Physics, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania;
| | - Maricel Agop
- Department of Physics, “Gheorghe Asachi” Technical University of Iasi, 700050 Iasi, Romania;
- Romanian Scientists Academy, 050094 Bucharest, Romania
| | - Cristian Constantin Volovat
- Department of Radiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.C.V.); (C.L.U.)
| | - Corina Lupascu Ursulescu
- Department of Radiology, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (C.C.V.); (C.L.U.)
| | | | - Simona Ruxandra Volovat
- Department of Medical Oncology-Radiotherapy, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Str, 700115 Iasi, Romania;
| |
Collapse
|
43
|
Davodabadi F, Farasati Far B, Sargazi S, Fatemeh Sajjadi S, Fathi-Karkan S, Mirinejad S, Ghotekar S, Sargazi S, Rahman MM. Nanomaterials-Based Targeting of Long Non-Coding RNAs in Cancer: A Cutting-Edge Review of Current Trends. ChemMedChem 2024; 19:e202300528. [PMID: 38267373 DOI: 10.1002/cmdc.202300528] [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: 09/30/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 01/26/2024]
Abstract
This review article spotlights the burgeoning potential of using nanotherapeutic strategies to target long non-coding RNAs (lncRNAs) in cancer cells. This updated discourse underlines the prominent role of lncRNAs in instigating cancer, facilitating its progression, and metastasis, validating lncRNAs' potential for being effective diagnostic biomarkers and therapeutic targets. The manuscript offers an in-depth examination of different strategies presently employed to modulate lncRNA expression and function for therapeutic purposes. Among these strategies, Antisense Oligonucleotides (ASOs), RNA interference (RNAi) technologies, and the innovative clustered regularly interspaced short palindromic repeats (CRISPR)-based gene editing tools garner noteworthy mention. A significant section of the review is dedicated to nanocarriers and their crucial role in drug delivery. These nanocarriers' efficiency in targeting lncRNAs in varied types of cancers is elaborated upon, validating the importance of targeted therapy. The manuscript culminates by reaffirming the promising prospects of targeting lncRNAs to enhance the accuracy of cancer diagnosis and improve treatment efficacy. Consequently, new paths are opened to more research and innovation in employing nanotherapeutic approaches against lncRNAs in cancer cells. Thus, this comprehensive manuscript serves as a valuable resource that underscores the vital role of lncRNAs and the various nano-strategies for targeting them in cancer treatment. Future research should also focus on unraveling the complex regulatory networks involving lncRNAs and identifying fundamental functional interactions to refine therapeutic strategies targeting lncRNAs in cancer.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran
| | - Bahareh Farasati Far
- Department of Chemistry, Iran University of Science and Technology, Tehran, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Seyedeh Fatemeh Sajjadi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| | - Sonia Fathi-Karkan
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 9453155166, Iran
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 9414974877, Iran
| | - Shekoufeh Mirinejad
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Suresh Ghotekar
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, 603103, Tamil Nadu, India
| | - Sara Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohammed M Rahman
- Center of Excellence for Advanced Materials Research (CEAMR) & Department of Chemistry, Faculty of Science, King Abdulaziz University, Jeddah, 21589, Saudi Arabia
| |
Collapse
|
44
|
Nisha R, Kumar P, Mishra N, Maurya P, Ahmad S, Singh N, Saraf SA. Appraisal of folate functionalized bosutinib cubosomes against hepatic cancer cells: In-vitro, In-silico, and in-vivo pharmacokinetic study. Int J Pharm 2024; 654:123975. [PMID: 38452833 DOI: 10.1016/j.ijpharm.2024.123975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 03/09/2024]
Abstract
Targeted therapies enhance the efficacy of tumour screening and management while lowering side effects. Multiple tumours, including liver cancer, exhibit elevated levels of folate receptor expression. This research attempted to develop surface-functionalised bosutinib cubosomes against hepatocellular carcinoma. The novelty of this work is the anti-hepatic action of bosutinib (BST) and folic acid-modified bosutinib cubosomes (BSTMF) established through proto-oncogene tyrosine-protein kinase (SrC)/ focal adhesion kinase(FAK), reactive oxygen species (ROS), mitochondrial membrane potential (MMP), and cell cytotoxicity. Later, the in-vivo pharmacokinetics of BSTMF were determined for the first time. The strong affinity of folic acid (FA) for folate receptors allows BSTMF to enter cells via FA receptor-mediated endocytosis. The particle size of the prepared BSTMF was 188.5 ± 2.25 nm, and its zeta potential was -20.19 ± 2.01 mV, an encapsulation efficiency of 90.31 ± 3.15 %, and a drug release rate of 76.70 ± 2.10 % for 48 h. The surface architecture of BSTMF was identified using transmission electron microscopy (TEM) and Atomic force microscopy (AFM). Cell-line studies demonstrated that BSTMF substantially lowered the viability of Hep G2 cells compared to BST and bosutinib-loaded cubosomes (BSTF). BSTMF demonstrated an elevated BST concentration in tumour tissue than in other organs and also displayed superior pharmacokinetics, implying that they hold potential against hepatic cancers. This is the first study to show that BSTMF may be effective against liver cancer by targeting folate receptors and triggering SrC/FAK-dependent apoptotic pathways. Multiple parameters demonstrated that BSTMF enhanced anticancer targeting, therapeutic efficacy, and safety in NDEA-induced hepatocellular carcinoma.
Collapse
Affiliation(s)
- Raquibun Nisha
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Pranesh Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India; Department of Pharmacology, Institute of Pharmaceutical Sciences, University of Lucknow, Lucknow, 226031, India
| | - Nidhi Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Priyanka Maurya
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India; Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Sector II, Dr Akhilesh Das Nagar, Faizabad Road, Lucknow, 226028, India
| | - Shakir Ahmad
- Department of Chemistry, Aligarh Muslim University, Aligarh, 202002, India
| | - Neelu Singh
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Shubhini A Saraf
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, India; National Institute of Pharmaceutical Education and Research (NIPER), Raebareli, Transit Campus: NIPER-Raebareli, Sarojini Nagar, Near CRPF Base Camp, Lucknow 226002, India.
| |
Collapse
|
45
|
Rich AL, Lin P, Gamazon ER, Zinkel SS. The broad impact of cell death genes on the human disease phenome. Cell Death Dis 2024; 15:251. [PMID: 38589365 PMCID: PMC11002008 DOI: 10.1038/s41419-024-06632-7] [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: 09/13/2023] [Revised: 03/09/2024] [Accepted: 03/22/2024] [Indexed: 04/10/2024]
Abstract
Cell death mediated by genetically defined signaling pathways influences the health and dynamics of all tissues, however the tissue specificity of cell death pathways and the relationships between these pathways and human disease are not well understood. We analyzed the expression profiles of an array of 44 cell death genes involved in apoptosis, necroptosis, and pyroptosis cell death pathways across 49 human tissues from GTEx, to elucidate the landscape of cell death gene expression across human tissues, and the relationship between tissue-specific genetically determined expression and the human phenome. We uncovered unique cell death gene expression profiles across tissue types, suggesting there are physiologically distinct cell death programs in different tissues. Using summary statistics-based transcriptome wide association studies (TWAS) on human traits in the UK Biobank (n ~ 500,000), we evaluated 513 traits encompassing ICD-10 defined diagnoses and laboratory-derived traits. Our analysis revealed hundreds of significant (FDR < 0.05) associations between genetically regulated cell death gene expression and an array of human phenotypes encompassing both clinical diagnoses and hematologic parameters, which were independently validated in another large-scale DNA biobank (BioVU) at Vanderbilt University Medical Center (n = 94,474) with matching phenotypes. Cell death genes were highly enriched for significant associations with blood traits versus non-cell-death genes, with apoptosis-associated genes enriched for leukocyte and platelet traits. Our findings are also concordant with independently published studies (e.g. associations between BCL2L11/BIM expression and platelet & lymphocyte counts). Overall, these results suggest that cell death genes play distinct roles in their contribution to human phenotypes, and that cell death genes influence a diverse array of human traits.
Collapse
Affiliation(s)
- Abigail L Rich
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Phillip Lin
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric R Gamazon
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Sandra S Zinkel
- Department of Pathology, Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
46
|
Saeinasab M, Atlasi Y, M Matin M. Functional role of lncRNAs in gastrointestinal malignancies: the peculiar case of small nucleolar RNA host gene family. FEBS J 2024; 291:1353-1385. [PMID: 36282516 DOI: 10.1111/febs.16668] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/18/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
Long noncoding RNAs (lncRNAs) play crucial roles in normal physiology and are often de-regulated in disease states such as cancer. Recently, a class of lncRNAs referred to as the small nucleolar RNA host gene (SNHG) family have emerged as important players in tumourigenesis. Here, we discuss new findings describing the role of SNHGs in gastrointestinal tumours and summarize the three main functions by which these lncRNAs promote carcinogenesis, namely: competing with endogenous RNAs, modulating protein function, and regulating epigenetic marking. Furthermore, we discuss how SNHGs participate in different hallmarks of cancer, and how this class of lncRNAs may serve as potential biomarkers in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Morvarid Saeinasab
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Iran
| | - Yaser Atlasi
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, UK
| | - Maryam M Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Iran
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Iran
| |
Collapse
|
47
|
Dos Santos B, Bion MC, Goujon-Svrzic M, Maher P, Dafre AL. REAP+: A single preparation for rapid isolation of nuclei, cytoplasm, and mitochondria. Anal Biochem 2024; 687:115445. [PMID: 38135241 PMCID: PMC10843687 DOI: 10.1016/j.ab.2023.115445] [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: 10/27/2023] [Revised: 12/03/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
REAP+ is an enhanced version of the rapid, efficient, and practical (REAP) method designed for the isolation of nuclear fractions. This improved version, REAP+, enables fast and effective extraction of mitochondria, cytoplasm, and nuclei. The mechanical cell disruption process has been optimized to cerebral tissues, snap-frozen liver, and HT22 cells with remarkable fraction enrichment. REAP+ is well-suited for samples containing minimal protein quantities, such as mouse hippocampal slices. The method was validated by Western blot and marker enzyme activities, such as LDH and G6PDH for the cytoplasmic fraction and succinate dehydrogenase and cytochrome c oxidase for the mitochondrial fraction. One of the outstanding features of this method is its rapid execution, yielding fractions within 15 min, allowing for simultaneous preparation of multiple samples. In essence, REAP+ emerges as a swift, efficient, and practical technique for the concurrent isolation of nuclei, cytoplasm, and mitochondria from various cell types and tissues. The method would be suitable to study the multicompartment translocation of proteins, such as metabolic enzymes and transcription factors migrating from cytosol to the mitochondria and nuclei. Moreover, its compatibility with small samples, such as hippocampal slices, and its potential applicability to human biopsies, highlights the potential application in medical research.
Collapse
Affiliation(s)
- Barbara Dos Santos
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil; Post-Graduation Program in Biochemistry, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil.
| | - Monique Coelho Bion
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil; Post-Graduation Program in Cell Biology and Development, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil.
| | - Marie Goujon-Svrzic
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, CA, 92037, La Jolla, United States.
| | - Pamela Maher
- Cellular Neurobiology Laboratory, Salk Institute for Biological Studies, CA, 92037, La Jolla, United States.
| | - Alcir Luiz Dafre
- Department of Biochemistry, Federal University of Santa Catarina, 88040-900, Florianópolis, SC, Brazil.
| |
Collapse
|
48
|
Zablon F, Desai P, Dellinger K, Aravamudhan S. Cellular and Exosomal MicroRNAs: Emerging Clinical Relevance as Targets for Breast Cancer Diagnosis and Prognosis. Adv Biol (Weinh) 2024; 8:e2300532. [PMID: 38258348 PMCID: PMC11198028 DOI: 10.1002/adbi.202300532] [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: 10/02/2023] [Revised: 12/26/2023] [Indexed: 01/24/2024]
Abstract
Breast cancer accounts for the highest cancer cases globally, with 12% of occurrences progressing to metastatic breast cancer with a low survival rate and limited effective early intervention strategies augmented by late diagnosis. Moreover, a low concentration of prognostic and predictive markers hinders disease monitoring. Circulating and exosomal microRNAs (miRNAs) have recently shown a considerable interplay in breast cancer, standing out as effective diagnostic and prognostic markers. The primary functions are as gene regulatory agents at the genetic and epigenetic levels. An array of dysregulated miRNAs stimulates cancer-promoting mechanisms, activating oncogenes and controlling tumor-suppressing genes and mechanisms. Exosomes are vastly studied extracellular vesicles, carrying, and transporting cargo, including noncoding RNAs with premier roles in oncogenesis. Translocation of miRNAs from the circulation to exosomes, with RNA-binding proteins in stress-induced conditions, has shown significant cooperation in function to promote breast cancer. This review examines cellular and exosomal miRNA biogenesis and loading, the clinical implications of their dysregulation, their function in diagnosis, prognosis, and prediction of breast cancer, and in regulating cancer signaling pathways. The influence of cellular and exosomal miRNAs presents clinical significance on breast cancer diagnosis, subtyping, staging, prediction, and disease monitoring during treatment, hence a potent marker for breast cancer.
Collapse
Affiliation(s)
- Faith Zablon
- Joint School of Nanoscience and Nanoengineering, North Carolina, A & T State University, 2904 E. Gate City Blvd, Greensboro, NC-27401
| | - Parth Desai
- University of North Carolina, Greensboro, 2904 E. Gate City Blvd, Greensboro, NC-27401
| | - Kristen Dellinger
- Joint School of Nanoscience and Nanoengineering, North Carolina, A & T State University, 2904 E. Gate City Blvd, Greensboro, NC-27401
| | - Shyam Aravamudhan
- Joint School of Nanoscience and Nanoengineering, North Carolina, A & T State University, 2904 E. Gate City Blvd, Greensboro, NC-27401
| |
Collapse
|
49
|
Chan KI, Zhang S, Li G, Xu Y, Cui L, Wang Y, Su H, Tan W, Zhong Z. MYC Oncogene: A Druggable Target for Treating Cancers with Natural Products. Aging Dis 2024; 15:640-697. [PMID: 37450923 PMCID: PMC10917530 DOI: 10.14336/ad.2023.0520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/20/2023] [Indexed: 07/18/2023] Open
Abstract
Various diseases, including cancers, age-associated disorders, and acute liver failure, have been linked to the oncogene, MYC. Animal testing and clinical trials have shown that sustained tumor volume reduction can be achieved when MYC is inactivated, and different combinations of therapeutic agents including MYC inhibitors are currently being developed. In this review, we first provide a summary of the multiple biological functions of the MYC oncoprotein in cancer treatment, highlighting that the equilibrium points of the MYC/MAX, MIZ1/MYC/MAX, and MAD (MNT)/MAX complexes have further potential in cancer treatment that could be used to restrain MYC oncogene expression and its functions in tumorigenesis. We also discuss the multifunctional capacity of MYC in various cellular cancer processes, including its influences on immune response, metabolism, cell cycle, apoptosis, autophagy, pyroptosis, metastasis, angiogenesis, multidrug resistance, and intestinal flora. Moreover, we summarize the MYC therapy patent landscape and emphasize the potential of MYC as a druggable target, using herbal medicine modulators. Finally, we describe pending challenges and future perspectives in biomedical research, involving the development of therapeutic approaches to modulate MYC or its targeted genes. Patients with cancers driven by MYC signaling may benefit from therapies targeting these pathways, which could delay cancerous growth and recover antitumor immune responses.
Collapse
Affiliation(s)
- Ka Iong Chan
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Siyuan Zhang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Guodong Li
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Yida Xu
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Liao Cui
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang 524000, China
| | - Yitao Wang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Huanxing Su
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| |
Collapse
|
50
|
Luo NF, Li JL, Lv J, Chen FK, Li YN, Tang M, Liu PJ. Role of sodium/iodide symporter overexpression in inhibiting thyroid cancer cell invasion and stem cell maintenance by inhibiting the β-catenin/LEF-1 pathway. Heliyon 2024; 10:e27840. [PMID: 38545139 PMCID: PMC10965522 DOI: 10.1016/j.heliyon.2024.e27840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 03/07/2024] [Accepted: 03/07/2024] [Indexed: 11/11/2024] Open
Abstract
Background In thyroid cancers, a reduction in the expression of the sodium/iodide symporter (NIS) is observed concomitant with a diminution in cancer cell differentiation. The β-catenin/LEF-1 pathway emerges as a crucial regulatory pathway influencing the functional expression of NIS in human thyroid cancer cells. Further research is required to comprehensively elucidate the role of NIS overexpression in impeding the progression of thyroid cancer cells. Methods Human papillary thyroid carcinoma (PTC) cell lines, specifically PTC-1 and KTC-1, were subjected to Scratch and Transwell assays, colony formation, and tumor sphere formation tests to investigate invasion and migration, focusing on the impact of NIS overexpression. The assessment involved the use of western blot to analyze the expression levels of β-catenin, NIS, CD133, SRY-related HMG box2 (Sox2), lymphoid enhancer-binding factor 1 (LEF-1), NANOG, octamer-binding transcription factor 4 (Oct4), aldehyde dehydrogenase 1 family, member A1 (ALDH1A1), and epithelial cellular adhesion molecule (EpCAM). Statistical analysis was conducted using SPSS version 20.0, and the graphs were developed using GraphPad Prism 7 (GraphPad Software, Inc.). Results Our observations revealed that Nthy-ori-3-1 cell lines exhibited notably higher average expression levels of NIS, yet significantly lower levels of LEF-1 and β-catenin compared to PTC-1 and KTC-1 cell lines. Furthermore, the overexpression of β-catenin resulted in reduced binding of LEF-1 to NIF promotion but concurrently increased the expression of NIS. The downregulation of NIS markedly enhanced the expression of ALDH1A1, CD133, OCT4, Nanog, SOX2, and EpCam-all of which are targets within the Wnt/β-catenin signaling pathway. Conversely, the upregulation of NIS suppressed the expression of these proteins. Moreover, cells treated with β-catenin activators demonstrated an increased capability to form more spheroids and displayed heightened aggressiveness. Conversely, the NIS overexpression (OE) group exhibited suppressed abilities in invasion and colony formation. Conclusion Thyroid cancer cells exhibit diminished expression of NIS, and the invasion and maintenance of stem cells in thyroid cancer cells were hindered by NIS OE through the inhibition of the β-catenin/LEF-1 pathway. Further research is warranted to comprehensively assess this outcome, which holds promise as a potential targeted treatment for thyroid cancer.
Collapse
Affiliation(s)
- Nan-Fang Luo
- Department of Cardiac Function, The Cancer Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Jia-Li Li
- Department of Nuclear Medicine, The Cancer Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Juan Lv
- Department of Nuclear Medicine, The Cancer Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Fu-Kun Chen
- Department of Nuclear Medicine, The Cancer Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Ya-Nan Li
- Department of Nuclear Medicine, The Cancer Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| | - Ming Tang
- Department of Pathology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, 650032, China
| | - Peng-Jie Liu
- Department of Nuclear Medicine, The Cancer Hospital of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming, 650118, China
| |
Collapse
|