1
|
Fiołka MJ, Sofińska-Chmiel W, Procházková L, Mieszawska S, Dryglewska M, Skrzypiec K, Wydrych J. Morphological and environmental analysis of the glacier ice alga Ancylonema alaskanum. Sci Rep 2025; 15:18578. [PMID: 40425824 PMCID: PMC12117168 DOI: 10.1038/s41598-025-95754-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/24/2025] [Indexed: 05/29/2025] Open
Abstract
In the presented study, the cells of the glacial alga Ancylonema alaskanum collected in the Austrian Alps were analyzed. Algae were imaged both in their natural environment and in laboratory conditions using transmitted light and fluorescence microscopy. Using appropriate fluorochromes, the cell wall and cell organelles were studied. Oval nuclei located in the middle of the cell next to the chloroplasts and active mitochondria as well as lipid thylakoids of chloroplasts were imaged. Scanning electron microscopy showed that the surface of the algal cell wall was not significantly differentiated, and atomic force microscope imaging recorded little roughness. The SEM EDS analysis revealed that carbon, nitrogen, oxygen, and magnesium were the main components of the cells. It is worth emphasizing that the analyzed living algal cells were obtained directly from the glacier surface and demonstrated normal respiratory processes i.e. undisturbed physiological functions. Additionally, the mineral material accompanying the cells in their natural environment - fragments of the rock were imaged by Differential Interference Contrast microscopy and analyzed by Fourier Transform Infrared Spectroscopy. The study provides new data on the morphology and physicochemical characteristics of A. alaskanum, contributing to a more comprehensive characterization of their place in this harsh ecosystem.
Collapse
Affiliation(s)
- Marta J Fiołka
- Department of Immunobiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland.
| | - Weronika Sofińska-Chmiel
- Analytical Laboratory, Institute of Chemistry, Maria Curie-Skłodowska University , Lublin, Poland
| | - Lenka Procházková
- Department of Ecology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Sylwia Mieszawska
- Chair and Department of Medical Microbiology, Medical University of Lublin, Lublin, Poland
| | - Magdalena Dryglewska
- Department of Rheumatology and Connective Tissue Diseases, Medical University of Lublin, Lublin, Poland
| | - Krzysztof Skrzypiec
- Analytical Laboratory, Institute of Chemistry, Maria Curie-Skłodowska University , Lublin, Poland
| | - Jerzy Wydrych
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Lublin, Poland
| |
Collapse
|
2
|
Abd El-Kaream SA, Hassan NAM, Saleh HSA, Albahloul MAS, Khedr AM, El-Kholey SM. Microwave assisted drug delivery of titanium dioxide/rose Bengal conjugated chitosan nanoparticles for micro-photodynamic skin cancer treatment in vitro and in vivo. BMC Cancer 2025; 25:896. [PMID: 40389852 PMCID: PMC12090424 DOI: 10.1186/s12885-025-14285-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Micro-photodynamic therapy (MWPDT) combines photo-dynamic (PDT) and microwave-dynamic (MWDT) therapies with sensitizers, offers new avenues for cancer treatment. Despite the fact that novel sensitizers for MWPDT have been successfully synthesized, only a few are being employed effectively. The low tumor-targeting specificity, inability to transport sensitizer's deeper intratumorally, and deteriorating tumor microenvironment all restrict their anti-tumor efficacy. The current work was done aiming at microwave assisted drug delivery of titanium dioxide / rose Bengal conjugated chitosan nanoparticles (TiO2/RB@CSNP) for micro- photo-dynamic skin cancer (SKCA) treatment in vitro and in vivo as activated cancer treatment up-to-date modality. MATERIALS AND METHODS The study was conducted in vitro on human SKCA cells (A-375) and the study protocol application groups in vivo on Swiss albino mice treated with 7,12-dimethylbenz[a]anthracene (DMBA)/croton oil only and were not received any treatment for inducing SKCA, and only after SKCA induction the study treatment protocol began, treatment was daily with TiO2/RB@CSNP as MWPDT sensitizer with or without exposure to laser (IRL) or microwave (MW) or a combination of them for 3 min for two weeks. RESULTS Revealed that CSNP can be employed as effective TiO2/RB delivery system that directly targets SKCA cells. Additionally TiO2/RB@CSNP is a promising MWPS for and when combined with MWPDT can be very effective in treatment of SKCA-A-375 in vitro (cell viability decreased in a dose-dependent basis, the cell cycle progression in G0/G1 was slowed down, and cell death was induced as evidenced by an increase in the population of Pre-G cells, an increase in early and late apoptosis and necrosis, and an increase in autophagic cell death) and DMBA/croton oil SKCA-induce mice in vivo (induced antiproliferative genes (caspase 3,9, p53, Bax, TNFalpha), suppressed antiapoptotic and antiangiogenic genes (Bcl2,VEGF respectively) effectively reducing the tumors growth and leading to cancer cell death as well as decreased oxidative stress (MDA), and ameliorated enzymatic and non-enzymatic antioxidants (SOD, GR, GPx, GST, CAT, GSH, TAC) as well as renal (urea, creatinine) and hepatic (ALT, AST) functions. This process could be attributed to MWPDT; microwave and/or photo-chemical TiO2/RB activation mechanism and antioxidant potential of non activated TiO2/RB as well. CONCLUSION The results indicate that TiO2/RB@CSNP has great promise as an innovative, effective delivery system for selective localized treatment of skin cancer that is activated by MWPDT.
Collapse
Affiliation(s)
- Samir Ali Abd El-Kaream
- Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | | | | | | | | | - Sohier Mahmoud El-Kholey
- Medical Biophysics Department, Medical Research Institute, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
3
|
Singh K, Gupta K, Shukla S, Kumari AP, Kumar A. Repurposing Oseltamivir Against C AG Repeat Mediated Toxicity in Huntington's Disease and Spinocerebellar Ataxia Using Cellular and Drosophila Model. ACS OMEGA 2025; 10:14980-14993. [PMID: 40290909 PMCID: PMC12019426 DOI: 10.1021/acsomega.4c10338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/12/2025] [Accepted: 02/18/2025] [Indexed: 04/30/2025]
Abstract
Huntington's disease (HD) and Spinocerebellar Ataxia (SCA) are debilitating neurological disorders triggered by the expansion of CAG sequences within the specific genes (HTT and ATXN, respectively). These are characterized as poly glutamine (polyQ) disorders, which are marked by widespread neurodegeneration and metabolic irregularities across systemic, cellular, and intracellular levels. This study aimed to identify small molecules that specifically interact with and target the toxic CAG repeat RNA. Here, we investigated the neuroprotective effects of Oseltamivir, an antiviral drug, against the HD and SCA-causing CAG repeats, through biophysical, cellular, and Drosophila model-based studies. Using a multidimensional approach encompassing biophysical techniques, cellular assays, and a Drosophila model, we explored Oseltamivir's interaction with toxic CAG repeat RNA. Our comprehensive analyses, including circular dichroism (CD), isothermal titration calorimetry (ITC), electrophoretic mobility shift assay (EMSA), and nuclear magnetic resonance (NMR) spectroscopy, demonstrated Oseltamivir's specific binding affinity for AA mismatches and its potential to mitigate the toxicity associated with polyQ aggregation. Moreover, the identified U.S. FDA-approved drug effectively mitigated polyQ-induced toxicity in both HD cells and the Drosophila model of the disease. The results obtained from this drug repurposing approach are indicative of the neuro-shielding role of Oseltamivir in HD and several SCAs, paving the way for its translation into clinical practice to benefit patients afflicted with these devastating diseases.
Collapse
Affiliation(s)
| | | | | | - Aditi Pramod Kumari
- Department of Biosciences
and Biomedical Engineering, Indian Institute
of Technology Indore, Simrol, Indore 453552, India
| | - Amit Kumar
- Department of Biosciences
and Biomedical Engineering, Indian Institute
of Technology Indore, Simrol, Indore 453552, India
| |
Collapse
|
4
|
Abd El-Kaream SA, Hamoda SAH, El Kholey SM, El-Sharkawy AM. Pulsed cavitation ultrasound assisted delivery of cardamom, pistacia and laurel encapsulated micelles nanoparticles for sono-photodynamic lymphoma in vitro and in vivo treatment. Lasers Med Sci 2025; 40:156. [PMID: 40126665 PMCID: PMC11933185 DOI: 10.1007/s10103-025-04387-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 02/26/2025] [Indexed: 03/26/2025]
Abstract
Sono-photodynamic therapy (SPDT) has attracted a lot of interest as a cutting-edge therapeutic strategy in the field of cancer treatment. The essential part of SPDT is the sensitizer, which under laser photon and pulsed cavitation ultrasound sono-irradiation may transform sono and photo- energy into cytotoxic molecules. Photon absorption, targeting, penetration, and oxygen dependence remain challenges in sono- -photosensitizer (SPs) design. Rapid advancements in material science have prompted the creation of several SPs that create cytotoxic species with great selectivity, safety, and noninvasiveness for the treatment of tumors. The current study aims to provide an advanced method of activated cancer treatment by using pulsed cavitation to assist the delivery of cardamom, pistacia and laurel conjugated micelles nanoparticles (CPL-Micelle NP) for the sono-photodynamic lymphoma in vivo and in vitro treatment. Human lymphoma cells (U-937) were used in the in vitro study, and the in vivo application groups of the study protocol were Swiss albino mice treated with 9,10-Dimethyl-1,2-Benzanthracene (DMBA) only; they were not given any treatment to induce lymphoma. The study treatment protocol started only after lymphoma induction, and involved daily administration of CPL-Micelle NP as SPDT sensitizer whether or not to be exposed to photo- (IRL) or sono- (US) or a combination of them for three minutes for a period of two weeks. Indicated that Micelle NP is a useful CPL delivery mechanism that targets lymphoma cells directly. Furthermore, CPL-Micelle NP is a promising SPS that, when used in conjunction with SPDT, can be very effective in in vitro treating lymphoma-U-937 (in a dose-dependent manner cell viability declined, an increase in the cells population during the S and G2/M phases indicates that the cell cycle was arrested, and an increase in cell population in the Pre-G, autophagic cell death, as well as necrosis and early and late apoptosis, indicate that cell death was induced) and DMBA-Lymphoma-induced mice in vivo (induced antiproliferative genes, repressed antiangiogenic and antiapoptotic genes), successfully slowing the growth of tumors and even killing cancer cells, as well as lowering oxidative stress malondialdehyde (MDA), improving the functions of the kidneys, liver, and enzymatic and non enzymatic antioxidants. SPDT, the photo- or sono- chemical CPL activation mechanism, and the antioxidant capacity of non-activated CPL can all be linked to this process. On the bases of the findings, CPL-Micelle NP shows a great promise as a novel, efficient selective delivery system for localized SPDT-activated lymphoma treatment.
Collapse
|
5
|
Sumorek-Wiadro J, Kapral-Piotrowska J, Zając A, Maciejczyk A, Hułas-Stasiak M, Skalicka-Woźniak K, Rzeski W, Pawlikowska-Pawlęga B, Jakubowicz-Gil J. Proapoptotic and antimigration properties of osthole in combination with LY294002 against human glioma cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:3147-3161. [PMID: 39352533 PMCID: PMC11919984 DOI: 10.1007/s00210-024-03424-w] [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: 08/09/2024] [Accepted: 08/30/2024] [Indexed: 03/19/2025]
Abstract
Anaplastic astrocytoma and glioblastoma multiforme are infiltrating and vascularized gliomas with a high degree of chemoresistance and metastasis. Our previous studies have shown that osthole may be of great importance in the treatment of gliomas. Therefore, in this work, for the first time, coumarin was used in combination with LY294002-an inhibitor of the PI3K-Akt/PKB-mTOR pathway, which is overly active in gliomas. MOGGCCM and T98G cells were incubated with osthole and LY294002, alone and in combination. Staining with specific fluorochromes was used to visualize cell death and the scratch test to assess the migration. The level of proteins was estimated by immunoblotting. Forming protrusions were visualized by SEM, and immunocytochemistry was used to determine the localization of proteins. Additionally, the expression of Bcl-2, beclin 1 and Raf kinase was silenced using specific siRNA. The obtained results showed that osthole in combination with LY294092 effectively inhibited the migration of glioma cells by reducing the level of metaloproteinases and Rho family proteins, as well as decreasing the level of N-cadherin. In addition, the combination of compounds induced apoptosis. New combination of compounds shows a high pro-apoptotic potential and also inhibits the migration of gliomas cells.
Collapse
Affiliation(s)
- Joanna Sumorek-Wiadro
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland.
| | - Justyna Kapral-Piotrowska
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Adrian Zając
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Aleksandra Maciejczyk
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Monika Hułas-Stasiak
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Krystyna Skalicka-Woźniak
- Independent Laboratory of Natural Products, Medical University of Lublin, Chodzki 1, 20-093, Lublin, Poland
| | - Wojciech Rzeski
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
- Department of Medical Biology, Institute of Rural Health, Institute of Agricultural Medicine, Jaczewskiego 2, 20-950, Lublin, Poland
| | - Bożena Pawlikowska-Pawlęga
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
| | - Joanna Jakubowicz-Gil
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Sklodowska University, Akademicka 19, 20-033, Lublin, Poland
| |
Collapse
|
6
|
Dey S, Nayak AK, Rajaram H, Das S. Exploitative stress within Bacillus subtilis biofilm determines the spatial distribution of pleomorphic cells. Microbiol Res 2025; 292:128034. [PMID: 39729737 DOI: 10.1016/j.micres.2024.128034] [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: 07/16/2024] [Revised: 11/04/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024]
Abstract
Bacteria commonly live in a spatially organized biofilm assemblage. The metabolic activity inside the biofilm leads to segmented physiological microenvironments. In nature, bacteria possess several pleomorphic forms to withstand certain ecological alterations. We hypothesized that pleomorphism also exists within the biofilm, which can be considered as the fundamental niche for bacteria. We report a distinct pattern of cell size variation throughout the biofilm of Bacillus subtilis. Cell size heterogeneity was observed in biofilm development, wherein the frequency of long cells is higher in outer regions, whereas lower in inner regions. Moreover, compared to planktonic cells, bacteria in the biofilm mode reduce their geometric ratio from 8.34 to 3.69 and 2.65 in the outer and inner regions, respectively. There were no significant differences observed in nutrient diffusion from the outer to the inner region, and more than 73 % of cells in the inner region were viable. However, the inner and middle regions were more acidic than the outer of the biofilm. Conclusively, growth rate-independent cell size reduction at low pH suggests that the resulting phenotype switching within biofilm was observed due to the pH gradient of neutral to acidic from the outer to the core of the biofilm. This gradient of H+ ions concentration may create exploitative stress within the biofilm, which could favor specific pleomorphic cells to thrive in their specialized niches. By understanding the cell size variation in response to the local environment, we propose a model of biofilm formation by pleomorphic cells.
Collapse
Affiliation(s)
- Sumon Dey
- Laboratory of Environmental Microbiology and Ecology (LEnME), Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Ankit Kumar Nayak
- Laboratory of Environmental Microbiology and Ecology (LEnME), Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Hema Rajaram
- Molecular Biology Division, Bhabha Atomic Research Institute, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai 400094, India
| | - Surajit Das
- Laboratory of Environmental Microbiology and Ecology (LEnME), Department of Life Science, National Institute of Technology, Rourkela, Odisha 769008, India.
| |
Collapse
|
7
|
Menpadi N, Chandra P, Dubey VK. Molecular Mechanisms of Cell Death in Leishmania donovani Induced by Selected Steroidal Alkaloids. J Basic Microbiol 2025; 65:e2400655. [PMID: 39604153 DOI: 10.1002/jobm.202400655] [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: 09/30/2024] [Revised: 10/30/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024]
Abstract
We have earlier reported novel anti-leishmanial molecules, veratramine and hupehenine, targeting dephospho-coenzyme A kinase of the parasite. In our current investigation, we assessed the efficacy of these two steroidal alkaloids, veratramine and hupehenine, in combating the parasite. Contrary to expectations, our study did not detect the typical signs of apoptosis such as mitochondrial membrane potential loss and phosphatidylserine externalization. Instead, we observed a notable increase in acidic organelle formation, suggesting a pro-survival response in promastigotes. Through diverse flow cytometric analyses and imaging methods, we conclude that the parasitic death induced by these natural compounds does not follow the apoptosis pathway but likely involves autophagy. This discovery marks the first instance of autophagy-mediated cell death in Leishmania donovani triggered by veratramine and hupehenine.
Collapse
Affiliation(s)
- Naveena Menpadi
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Pranjal Chandra
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Vikash Kumar Dubey
- School of Biochemical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| |
Collapse
|
8
|
Lai GY, Lee YC, Weng HJ, Lai KH, Hsiang MC, Hsu KY, Liao CP. Discoidin domain receptor inhibitor DDR1-IN-1 induces autophagy and necroptotic cell death in malignant peripheral nerve sheath tumor. Cell Death Discov 2025; 11:83. [PMID: 40025071 PMCID: PMC11873111 DOI: 10.1038/s41420-025-02367-2] [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: 07/30/2024] [Revised: 01/14/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025] Open
Abstract
Malignant peripheral nerve sheath tumor (MPNST) is a soft tissue sarcoma commonly associated with the tumor-predisposition disorder neurofibromatosis 1. The extracellular matrix collagens contribute to many fibrotic tumors; however, the role of collagen signaling in MPNST was unclear. This study investigated the effects of blocking the interaction between collagens and their receptors in MPNST. We first analyzed the expressions of collagen family proteins in MPNSTs and found an overall increase compared to neurofibroma. Treatment of DDR1-IN-1, a small molecule inhibitor for the collagen receptor discoidin domain receptor, induced a robust MPNST cell death, highlighting the dependence of MPNST survival on collagen signaling. DDR1-IN-1 induced MPNST cell death by activating autophagy and necroptosis signaling. Treatment of necroptosis inhibitors necrostatin-1 or necrosulfonamide reduced the numbers of DDR1-IN-1-induced necrotic cells and autolysosomes, suggesting that the autophagic process depends on necroptosis activation. Combinations of DDR1-IN-1 with other anti-MPNST agents revealed synergistic activities against MPNST. In summary, this study discovered a critical MPNST death signaling induced by the small molecule DDR1-IN-1, which might shed light on future MPNST therapeutic strategies.
Collapse
Affiliation(s)
- Guan-Yi Lai
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Yu-Cheng Lee
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Hao-Jui Weng
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Dermatology, Taipei Medical University-Shuang Ho Hospital, New Taipei City, 23561, Taiwan
- Department of Dermatology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kuei-Hung Lai
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- PhD Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan
| | - Min-Chen Hsiang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Kai-Yu Hsu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chung-Ping Liao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- International Ph.D. Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Cancer Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan.
- Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, 11696, Taiwan.
| |
Collapse
|
9
|
Cavada B, Pinto-Junior VR, Lima FEO, Ferreira VMS, Oliveira MV, Osterne VJS, Sartori N, Santos ACD, Leal RB, Nascimento KS. Crystallographic Structure and Antiglioma Potential of Centrolobium microchaete Seed Lectin. ACS OMEGA 2025; 10:4686-4698. [PMID: 39959084 PMCID: PMC11822720 DOI: 10.1021/acsomega.4c09145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/06/2025] [Accepted: 01/14/2025] [Indexed: 02/18/2025]
Abstract
The genus Centrolobium comprises species of Neotropical trees with seeds that possess medicinal and bioactive applications. Lectins from this genus exhibit anti-inflammatory and immunomodulatory effects, influencing the activation of the immune system. This study focuses on characterizing the structure and carbohydrate-binding properties of the lectin from Centrolobium microchaete (CML) and evaluating its potential against gliomas. The structure of the lectin in complex with methyl-mannose-α1,3-mannose (MDM) was resolved using X-ray crystallography at 1.3 Å resolution, with its interactions further analyzed through molecular dynamics simulations. Structurally, CML adopts a β-sandwich motif and assembles into canonical dimers. In vitro assays revealed that CML reduced the viability of C6 glioma cells, although only at high concentrations, without impacting cell migration or morphology. CML activated autophagic processes, albeit with lower efficacy compared with other mannose-specific lectins. The limited antiglioma activity of CML may be linked to its inability to form tetramers and unusual specificity toward asymmetric glycans, both crucial features for interactions with cellular glycans and the activation of signaling pathways. This study represents the first investigation of the antiglioma potential of a mannose-specific lectin from the Dalbergieae tribe, highlighting both its structural characteristics and functional limitations.
Collapse
Affiliation(s)
- Benildo
Sousa Cavada
- Department
of Biochemistry and Molecular Biology, BioMolLab, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
| | - Vanir Reis Pinto-Junior
- Department
of Biochemistry and Molecular Biology, BioMolLab, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
| | | | - Valeria Maria Sousa Ferreira
- Department
of Biochemistry and Molecular Biology, BioMolLab, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
| | - Messias Vital Oliveira
- Department
of Biochemistry and Molecular Biology, BioMolLab, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
| | - Vinicius Jose Silva Osterne
- Department
of Biochemistry and Molecular Biology, BioMolLab, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
- Laboratory
of Biochemistry and Glycobiology, Department of Biotechnology, Ghent University, 9000 Ghent, Belgium
| | - Nicole Sartori
- Department
of Biochemistry and Postgraduate Program in Biochemistry, Center for
Biological Sciences, University Campus, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Ana Carolina dos Santos
- Department
of Biochemistry and Postgraduate Program in Biochemistry, Center for
Biological Sciences, University Campus, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Rodrigo Bainy Leal
- Department
of Biochemistry and Postgraduate Program in Biochemistry, Center for
Biological Sciences, University Campus, Federal University of Santa Catarina, Florianópolis 88040-900, SC, Brazil
| | - Kyria Santiago Nascimento
- Department
of Biochemistry and Molecular Biology, BioMolLab, Federal University of Ceara, Fortaleza 60020-181, CE, Brazil
| |
Collapse
|
10
|
He M, Wu H, Xu T, Zhao Y, Wang Z, Liu Y. Fangchinoline eliminates intracellular Salmonella by enhancing lysosomal function via the AMPK-mTORC1-TFEB axis. J Adv Res 2025:S2090-1232(25)00034-7. [PMID: 39788287 DOI: 10.1016/j.jare.2025.01.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/19/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025] Open
Abstract
INTRODUCTION Salmonella, a foodborne zoonotic pathogen, is a significant cause of morbidity and mortality in animals and humans globally. With the prevalence of multidrug-resistant strains, Salmonellosis has become a formidable challenge. Host-directed therapy (HDT) has recently emerged as a promising anti-infective approach for treating intracellular bacterial infections. OBJECTIVES Plant-derived natural products, owing to their structural and functional diversity, are increasingly being explored and utilized as encouraging candidates for HDT compounds. This study aims to identify and screen natural compounds with potential as HDT for the treatment of intracellular Salmonella infections. METHODS A cell-based screening approach was deployed to identify natural compounds capable of mitigating the intracellular replication of S. enterica. Safety and efficacy of the candidate compounds were evaluated using multiple animal models. RNA sequencing, ELISA, and immunoblotting analyses were conducted to elucidate the underlying mechanisms of action. RESULTS Our results reveal that fangchinoline (FAN) effectively reduces S. enterica survival both in vitro and in vivo. Meanwhile, FAN also displays anti-infective activity against other intracellular pathogens, including multidrug-resistant isolates. A 14-day safety evaluation in mice showed no significant toxic or adverse effects from FAN administration. RNA sequencing analysis reveals an upregulation of lysosome pathways in S. enterica-infected cells treated with FAN. Mechanistic studies indicate that FAN increases acid lysosomal quantities and fosters autophagic response in Salmonella-infected cells via the AMPK-mTORC1-TFEB axis. In addition, FAN alleviates the inflammatory response in Salmonella-infected cells by inactivating the NF-κB pathway. CONCLUSION Our findings suggest that FAN represents a lead HDT compound for tackling recalcitrant infections caused by intracellular bacterial pathogens.
Collapse
Affiliation(s)
- Mengping He
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Huihui Wu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Tianqi Xu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yurong Zhao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhiqiang Wang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China.
| | - Yuan Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.
| |
Collapse
|
11
|
Onwah SS, Uzonna JE, Ghavami S. Assessment of Autophagy in Leishmania Parasites. Methods Mol Biol 2025; 2879:207-217. [PMID: 38441724 DOI: 10.1007/7651_2024_517] [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: 02/19/2025]
Abstract
Leishmaniasis is a neglected tropical disease caused by numerous species of Leishmania parasites, including Leishmania major. The parasite is transmitted by several species of sandfly vectors and infects myeloid cells leading to a myriad of inflammatory responses, immune dysregulations, and disease manifestations. Every cell undergoes autophagy, a self-regulated degradative process that permits the cells to recycle damaged or worn-out organelles in order to maintain cellular health and homeostasis. Studies have shown that Leishmania modulates their host cell autophagic machinery and there are indications that the parasite-specific autophagic processes may be valuable for parasite virulence and survival. However, the role of autophagy in Leishmania is inconclusive because of the limited tools available to study the Leishmania-specific autophagic machinery. Here, we describe methods to study and definitively confirm autophagy in Leishmania major. Transmission electron microscopy (TEM) allowed us to visualize Leishmania autophagosomes, especially those containing damaged mitochondrial content, as well as dividing mitochondria with ongoing fusion/fission processes. Flow cytometry enabled us to identify the amount of acridine orange dye accumulating in the acidic vacuolar compartments in Leishmania major by detecting fluorescence in the red laser when autophagic inhibitors or enhancers were included. These methods will advance studies that aim to understand autophagic regulation in Leishmania parasites that could provide insights into developing improved therapeutic targets against leishmaniasis.
Collapse
Affiliation(s)
- Somtochukwu S Onwah
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada.
| | - Jude E Uzonna
- Department of Immunology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Medical Microbiology and Infectious Diseases, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Department of Pathology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Saeid Ghavami
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada.
- Academy of Silesia, Faculty of Medicine, Katowice, Poland.
- Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB, Canada.
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB, Canada.
| |
Collapse
|
12
|
Luo C, Li X, Yan H, Guo Q, Liu J, Li Y. Iron oxide nanoparticles induce ferroptosis under mild oxidative stress in vitro. Sci Rep 2024; 14:31383. [PMID: 39733146 PMCID: PMC11682176 DOI: 10.1038/s41598-024-82917-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 12/10/2024] [Indexed: 12/30/2024] Open
Abstract
Iron oxide nanoparticles (IONPs) have the potential to be utilized in a multitude of fields, including biomedicine. Consequently, the potential health risks associated with their use must be carefully considered. Most biosafety evaluations of IONPs have focused on examining the impact of the material's distinctive physicochemical attributes. However, the specific attributes of individual cells are frequently disregarded, particularly under the oxidative stress conditions. This may result in an underestimation of potential risk and impede the clinical translation of IONPs. The present study thus sought to evaluate the potential cytotoxicity and underlying mechanisms of IONPs in a pathological state characterized by mild oxidative stress. A cell model of mild oxidative stress was initially established in vitro. Subsequently, a series of indicators, including cell viability, live/dead ratio, mitochondrial membrane potential, and oxidative damage, were measured to assess the cytotoxicity of IONPs. Finally, a series of ferroptosis regulators were used to elucidate the involvement of ferroptosis. Preincubation with IONPs resulted in a significant reduction in cellular viability, morphological degeneration, elevated numbers of dead cells, impaired mitochondrial function, and increased oxidative damage under mild oxidative stress conditions in vitro. The cytotoxic effects of IONPs under mild oxidative stress are largely dependent on ROS and iron ions and are strongly associated with ferroptosis, which is based on the effects of ferroptosis regulators. The present in vitro study indicated that IONPs are toxic to cells under mild oxidative stress, which is linked to ferroptosis.
Collapse
Affiliation(s)
- Cheng Luo
- School of Medicine, Yichun University, Yichun, 336000, China
| | - Xuying Li
- School of Medicine, Yichun University, Yichun, 336000, China
| | - Hongyang Yan
- School of Medicine, Yichun University, Yichun, 336000, China
| | - Qitao Guo
- School of Medicine, Yichun University, Yichun, 336000, China
| | - Jiarong Liu
- School of Medicine, Yichun University, Yichun, 336000, China
| | - Yan Li
- School of Medicine, Yichun University, Yichun, 336000, China.
| |
Collapse
|
13
|
Kim NY, Vishwanath D, Basappa S, Harish KK, Madegowda M, Rangappa KS, Basappa B, Ahn KS. Isoxazole based nucleosides induce autophagy through the production of ROS and the suppression of the β-catenin pathway in human colorectal carcinoma cells. Chem Biol Interact 2024; 404:111285. [PMID: 39442680 DOI: 10.1016/j.cbi.2024.111285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/17/2024] [Accepted: 10/20/2024] [Indexed: 10/25/2024]
Abstract
β-catenin is frequently implicated in signaling pathways that regulate autophagy, and the production of reactive oxygen species (ROS) has been linked to autophagy activation. Isoxazole-based nucleoside compounds have demonstrated anti-cancer properties. In this study, we report the identification of novel isoxazole-nucleosides as anti-tumor agents and their impact on autophagy in human colorectal carcinoma (CRC) cells. Among the ITP series, ITP-7 and ITP-9 (ITP-7/9) exhibited significant cytotoxicity compared to other compounds. Treatment with ITP-7/9 upregulated the expression of key autophagy-related proteins, including LC3 II, Atg7, and phosphorylated Beclin-1. Additionally, ITP-7/9 promoted the formation of LC3 II puncta and increased the number of AO-stained and MDC-stained cells, indicating enhanced autophagy. ROS levels were elevated following ITP-7/9 exposure, and treatment with N-acetyl l-cysteine (NAC), a ROS inhibitor, reduced the ITP-7/9-induced expression of LC3 II. Furthermore, ITP-7/9 inhibited β-catenin's role as a transcription factor, as observed in ICC assays. Moreover, cells with β-catenin gene deletion exhibited stronger autophagy when treated with ITP-7/9 compared to those treated with ITP-7/9 alone. These findings suggest that ITP-7/9 induces autophagy and promotes CRC cell death by downregulating β-catenin.
Collapse
Affiliation(s)
- Na Young Kim
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Divakar Vishwanath
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysuru, 570006, India
| | - Shreeja Basappa
- Department of Chemistry, BITS-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, 500078, India
| | - Keshav Kumar Harish
- Department of Studies in Physics, University of Mysore, Manasagangotri, Mysore, 570006, India
| | - Mahendra Madegowda
- Department of Studies in Physics, University of Mysore, Manasagangotri, Mysore, 570006, India
| | - Kanchugarakoppal S Rangappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysuru, 570006, India
| | - Basappa Basappa
- Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysuru, 570006, India.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul, 02447, Republic of Korea.
| |
Collapse
|
14
|
Živanović M, Selaković M, Pavić A, Selaković Ž, Šolaja B, Santibanez JF, Srdić-Rajić T. Unveiling the 4-aminoquinoline derivatives as potent agents against pancreatic ductal adenocarcinoma (PDAC) cell lines. Chem Biol Interact 2024; 404:111281. [PMID: 39428053 DOI: 10.1016/j.cbi.2024.111281] [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: 07/31/2024] [Revised: 10/04/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Common antimalarials such as artemisinins, chloroquine and their derivatives also possess potent anti-inflamantory, antiviral and anticancer properties. In the search for new therapeutics to combat difficult-to-treat pancreatic carcinomas, we unveiled that 4-aminoquinoline derivatives, with significant antiplasmodial properties and a great safety profile in vivo, have remarkable anticancer activity against pancreatic ductal adenocarcinoma (PDAC) and considerable efficacy in the xenograft model in vivo. The aim of the present study was to further investigate anticancer properties of these compounds in a drug-repurposing manner. The compounds showed profound cytotoxic effects at nanomolar to low micromolar concentration in 2D cultured cells (in vitro) and in the zebrafish PDAC xenograft model (in vivo). A deeper insight into their mechanisms of cytotoxic action showed these compounds induce apoptosis while increasing reactive oxygen species levels along with autophagy inhibition. Additional investigation of the autophagy modulation proved that tested quinoline derivatives cause P62 and LC3-II accumulation in PDAC cells alongside lysosomal alkalinization. Further, in vivo toxicity studies in the zebrafish model showed low toxicity without developmental side effects of the investigated 4-aminoquinolines, while the applied compounds effectively inhibited tumor growth and prevented the metastasis of xenografted pancreatic cells. Taken together, these results highlight the 4-aminoquinolines as privileged structures that ought to be investigated further for potential application in pancreatic carcinoma treatment.
Collapse
Affiliation(s)
- Marija Živanović
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia; Department of Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Dr. Subotića 4, 11129 Belgrade, Serbia
| | - Milica Selaković
- Innovative Centre of the Faculty of Chemistry in Belgrade, ltd., Studentski Trg 12-16, 11158, Belgrade, Serbia.
| | - Aleksandar Pavić
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Vojvode Stepe 444a, 11042, Belgrade, Serbia
| | - Života Selaković
- University of Belgrade - Faculty of Chemistry, Studentski Trg 12-16, 11158, Belgrade, Serbia
| | - Bogdan Šolaja
- Serbian Academy of Sciences and Arts, Knez Mihailova 35, 11158, Belgrade, Serbia
| | - Juan F Santibanez
- Department of Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Dr. Subotića 4, 11129 Belgrade, Serbia
| | - Tatjana Srdić-Rajić
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia.
| |
Collapse
|
15
|
Varga K, Paszternák A, Kovács V, Guczogi A, Sikur N, Patakfalvi D, Bagaméry F, Szökő É, Tábi T. Differential Cytoprotective Effect of Resveratrol and Its Derivatives: Focus on Antioxidant and Autophagy-Inducing Effects. Int J Mol Sci 2024; 25:11274. [PMID: 39457058 PMCID: PMC11509103 DOI: 10.3390/ijms252011274] [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: 09/21/2024] [Revised: 10/14/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
Numerous beneficial effects of resveratrol were reported; however, its pharmacological profile is contradictious. Previously, we have demonstrated that resveratrol has a dose-dependent cytoprotective effect and the essential role of autophagy induction was demonstrated. Resveratrol suffers from unfavorable pharmacokinetics, hindering its clinical use. Our aim was to study the cytoprotective effect of resveratrol derivatives to better understand structure-activity relationships that may facilitate the development of compounds with better druglike characteristics. Serum-deprivation-induced caspase activation, free radical generation, mitochondrial membrane depolarization and autophagy were detected in the presence of resveratrol analogs with different oxidation states on mouse embryonal fibroblasts. Distinct cytoprotective mechanisms of the examined compounds were revealed. Monomethyl resveratrol had similar potency to resveratrol (EC50: 85.3 vs. 84.2 μM); however, autophagy induction was not essential for its cytoprotective effect. Oxyresveratrol was found to be a strong antioxidant that can induce direct cytoprotection rather than autophagy. Trimethyl-resveratrol, lacking free hydroxyl groups, induced damage that was too significant and hardly compensated by the activation of cytoprotective machineries, and caspase activation was reduced by only 24.5%. Based on our results, methylation of resveratrol reduces its antioxidant activity, while autophagy induction can still contribute to its cytoprotective effect. The introduction of an additional hydroxyl group, however, augments the antioxidant properties, inducing cytoprotection without autophagy induction.
Collapse
Affiliation(s)
- Kamilla Varga
- Department of Pharmacodynamics, Semmelweis University, 4 Nagyvárad tér, H-1089 Budapest, Hungary; (K.V.); (A.P.); (N.S.); (F.B.); (É.S.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 26 Üllői út, H-1085 Budapest, Hungary
| | - Alexandra Paszternák
- Department of Pharmacodynamics, Semmelweis University, 4 Nagyvárad tér, H-1089 Budapest, Hungary; (K.V.); (A.P.); (N.S.); (F.B.); (É.S.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 26 Üllői út, H-1085 Budapest, Hungary
| | - Virág Kovács
- Department of Pharmacodynamics, Semmelweis University, 4 Nagyvárad tér, H-1089 Budapest, Hungary; (K.V.); (A.P.); (N.S.); (F.B.); (É.S.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 26 Üllői út, H-1085 Budapest, Hungary
| | - Annamária Guczogi
- Department of Pharmacodynamics, Semmelweis University, 4 Nagyvárad tér, H-1089 Budapest, Hungary; (K.V.); (A.P.); (N.S.); (F.B.); (É.S.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 26 Üllői út, H-1085 Budapest, Hungary
| | - Noémi Sikur
- Department of Pharmacodynamics, Semmelweis University, 4 Nagyvárad tér, H-1089 Budapest, Hungary; (K.V.); (A.P.); (N.S.); (F.B.); (É.S.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 26 Üllői út, H-1085 Budapest, Hungary
| | - Dimitrisz Patakfalvi
- Department of Pharmacodynamics, Semmelweis University, 4 Nagyvárad tér, H-1089 Budapest, Hungary; (K.V.); (A.P.); (N.S.); (F.B.); (É.S.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 26 Üllői út, H-1085 Budapest, Hungary
| | - Fruzsina Bagaméry
- Department of Pharmacodynamics, Semmelweis University, 4 Nagyvárad tér, H-1089 Budapest, Hungary; (K.V.); (A.P.); (N.S.); (F.B.); (É.S.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 26 Üllői út, H-1085 Budapest, Hungary
| | - Éva Szökő
- Department of Pharmacodynamics, Semmelweis University, 4 Nagyvárad tér, H-1089 Budapest, Hungary; (K.V.); (A.P.); (N.S.); (F.B.); (É.S.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 26 Üllői út, H-1085 Budapest, Hungary
| | - Tamás Tábi
- Department of Pharmacodynamics, Semmelweis University, 4 Nagyvárad tér, H-1089 Budapest, Hungary; (K.V.); (A.P.); (N.S.); (F.B.); (É.S.)
- Center for Pharmacology and Drug Research & Development, Semmelweis University, 26 Üllői út, H-1085 Budapest, Hungary
| |
Collapse
|
16
|
Dutta A, Chakraborty A, Ghosh T, Kumar A. 5-Fluorouracil induces apoptosis in nutritional deprived hepatocellular carcinoma through mitochondrial damage. Sci Rep 2024; 14:23387. [PMID: 39379402 PMCID: PMC11461840 DOI: 10.1038/s41598-024-73143-y] [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: 06/24/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024] Open
Abstract
5-Fluorouracil (5-FU) is the leading chemotherapeutic drug used to treat hepatocellular carcinoma, one of the major cancer diseases after atherosclerosis. Because of chemo-resistance, the success rate of treatment declines with time due to continuous drug exposure. Though autophagy induction is majorly responsible for acquired resistance, the exact role of this evolutionary conserved mechanism is unknown in cancer cell survival and suppression. The usual practice involves the combinatorial use of chemotherapeutic drugs with autophagy inhibitors like Chloroquine and Bafilomycin A, while neglecting the side effects caused by autophagy impairment in healthy cells. Starvation is a well-known physiological inducer of autophagy. In this study, by caloric modulation, we tried to circumvent the resistance imposed by prolonged drug exposure and investigated the effect of 5-FU in nutrient-sufficient and deficient conditions. Our findings show a substantial correlation between autophagy and increased cancer cell death in the presence of 5-FU, with negligible effects on normal cells. Experimental data revealed that nutritional deprivation augmented cell death in the presence of 5-FU through mitochondrial membrane damage and excessive reactive oxygen species (ROS) production, initiating apoptosis. Lipidation study also unveiled that under such combinatorial treatment cellular metabolism shifts from glucose to lipid biosynthesis. Overall, our experimental findings suggest that nutritional deprivation in combination with chemotherapeutic medication can be a new effective strategy to control hepatocellular carcinoma.
Collapse
Affiliation(s)
- Ankita Dutta
- Advanced Nanoscale Molecular Oncology Laboratory (ANMOL), Department of Biotechnology, University of North Bengal, Raja Rammohunpur, District - Darjeeling, 734013, Siliguri, West Bengal, India
| | - Anuja Chakraborty
- Advanced Nanoscale Molecular Oncology Laboratory (ANMOL), Department of Biotechnology, University of North Bengal, Raja Rammohunpur, District - Darjeeling, 734013, Siliguri, West Bengal, India
| | - Tulika Ghosh
- Advanced Nanoscale Molecular Oncology Laboratory (ANMOL), Department of Biotechnology, University of North Bengal, Raja Rammohunpur, District - Darjeeling, 734013, Siliguri, West Bengal, India
| | - Anoop Kumar
- Advanced Nanoscale Molecular Oncology Laboratory (ANMOL), Department of Biotechnology, University of North Bengal, Raja Rammohunpur, District - Darjeeling, 734013, Siliguri, West Bengal, India.
| |
Collapse
|
17
|
Dong X, Liu X, Lin D, Zhang L, Wu Y, Chang Y, Jin M, Huang G. Baicalin induces cell death of non-small cell lung cancer cells via MCOLN3-mediated lysosomal dysfunction and autophagy blockage. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155872. [PMID: 39096542 DOI: 10.1016/j.phymed.2024.155872] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/24/2024] [Accepted: 07/09/2024] [Indexed: 08/05/2024]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) accounts for 85 % of lung cancer, becoming the most mortality of all cancers globally. Blockage of autophagy in NSCLC represents a promising therapeutic strategy that inhibits angiogenesis and overcomes drug resistance. Natural ingredients in anti-tumor adjuvants are increasingly reported to promote cell death with less side effects and the potential to increase chemotherapeutic drugs sensitivity. Baicalin, a Scutellaria baicalensis-extracted flavonoid glycoside, is reported to induce death of NSCLC cells, however, its effects on autophagy in NSCLC cells remain unclear. PURPOSE This study aimed to investigate the effect of baicalin on autophagic flux in NSCLC cells, unraveling the underlying mechanism including potential target and its role in cell death of NSCLC cells. METHODS In vitro anti-cancer effects of baicalin were verified by evaluating proliferation, clone formation, cell cycle, and cell migration in three NSCLC cell lines (A549, H1299, and PC-9). In vivo anti-tumor efficacies of baicalin were evaluated in subcutaneous xenograft tumor model in nude mice. Autophagy characterization in NSCLC cells included autophagic marker detection by western blot and immunofluorescence staining, subcellular structure observation by TEM, lysosomal function by RNA-seq and fluorescence staining (LysoTracker®, LysoSensor®, and acridine orange). Based on RNA-seq and molecular biological verification using apoptotic, autophagic, and lysosomal inhibitors, potential target molecule of baicalin was verified via Ca2+ flux assay, MCOLN3 knockdown by shRNA, and virtual molecular docking. RESULTS Baicalin inhibited NSCLC cell proliferation and migration, and suppressed tumor growth in vivo. Baicalin blocked the autophagic flux via activating the membranal cation channel MCOLN3 of lysosome, which disrupted its Ca2+ balance and induced lysosome dysfunction, leading to failure of autolysosome degradation. The cytoplasmic Ca2+ imbalance further resulted in depolarization of mitochondrial membrane potentials and ROS accumulation in NSCLC cells, mediating autophagy-related apoptosis. CONCLUSION This study demonstrated that baicalin inhibited autolysosome degradation by activating MCOLN3, leading to dysfunction in lysosomal pH elevation, thereby inhibiting autophagy in NSCLC, leading to apoptotic death of NSCLC cells. These findings enriched the existing theories of cancer therapy based on autophagy inhibition and underlying mechanisms of flavonoids as antitumor agents, paving the way for their clinical application in future.
Collapse
Affiliation(s)
- Xian Dong
- School of Graduate, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai 201318, China; College of Medical Technology, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai 201318, China
| | - Xiyu Liu
- School of Graduate, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai 201318, China
| | - Dan Lin
- College of Medical Technology, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai 201318, China.
| | - Lian Zhang
- School of Graduate, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai 201318, China; Department of Radiology, Jiading Hospital of Traditional Chinese Medicine, 222 Bo Le Rd. Shanghai 201800, China
| | - Yue Wu
- Department of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 274 Middle Zhi Jiang Rd. Shanghai 200071, China
| | - Yuzhen Chang
- School of Graduate, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai 201318, China
| | - Mingming Jin
- School of Graduate, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai 201318, China.
| | - Gang Huang
- School of Graduate, Shanghai University of Traditional Chinese Medicine, 1200 Cai Lun Rd. Shanghai 201203, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, 279 Zhouzhu Highway, Shanghai 201318, China.
| |
Collapse
|
18
|
Amifuji M, Inagaki M, Yoshida M, Doi T, Tachikawa M. Characteristics of membrane transport, metabolism, and target protein binding of cyclic depsipeptide destruxin E in HeLa cells. Drug Metab Pharmacokinet 2024; 58:101028. [PMID: 39265438 DOI: 10.1016/j.dmpk.2024.101028] [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: 05/12/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 09/14/2024]
Abstract
Cyclic peptides have attracted attention as new modalities for drug development owing to their unique pharmacokinetic and pharmacodynamic properties. Destruxin E, a 19-membered cyclodepsipeptide, is a promising candidate drug for cancer therapy. The purpose of the present study was to clarify the molecular mechanisms underlying membrane transport, metabolism, and the binding for target molecules of destruxin E in human cervical carcinoma HeLa cells used as a model of cancer cells. The influx transport and the intracellular metabolism of destruxin E were non-saturable and saturable, respectively, at up to 10 μM. The intracellular amounts of destruxin E and destruxin E-diol after incubation of destruxin E with the cells significantly decreased at 4 °C compared to those at 37 °C. Destruxin E-diol, but not destruxin E, undergoes efflux transport out of cells via P-gp/MDR1/ABCB1 and BCRP/ABCG2. The epoxide hydrolase EPHX2 functions as a potent metabolizing enzyme that can convert the epoxide of destruxin E to the destruxin E-diol. Treatment with an EPHX2 inhibitor increased the intracellular destruxin E levels and enhanced the inhibitory activity of vacuolar type-H+ ATPase. These results suggest that epoxide hydrolase could be a regulatory factor for intracellular destruxin E levels and its pharmacological activity.
Collapse
Affiliation(s)
- Makoto Amifuji
- Graduate School of Pharmaceutical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima, 770-8505, Japan
| | - Mai Inagaki
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima, 770-8505, Japan
| | - Masahito Yoshida
- Faculty of Pure and Applied Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, Ibaraki, 305-8571, Japan
| | - Takayuki Doi
- Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Masanori Tachikawa
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Sho-machi, Tokushima, 770-8505, Japan.
| |
Collapse
|
19
|
Zhang C, Feng Y, Calderin JD, Balutowski A, Ahmed R, Knapp C, Fratti RA. Lysophospholipid headgroup size, and acyl chain length and saturation differentially affect vacuole acidification, Ca 2+ transport, and fusion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.27.615487. [PMID: 39386589 PMCID: PMC11463366 DOI: 10.1101/2024.09.27.615487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
SNARE-mediated membrane fusion is regulated by the lipid composition of the engaged bilayers. Lipid composition impacts fusion through direct protein lipid interactions or through modulating the physical properties of membranes at the site of contact, including the induction of positive curvature by lysophospholipids (LPLs). The degree of positive curvature induced is due to the length and saturation of the single acyl chain in addition to the size of the head group. Here we examined how yeast vacuole fusion and ion transport were differentially affected by changes in lysolipid properties. We found that lysophosphatidylcholine (LPC) with acyl chains containing 14-18 carbons all inhibited fusion with IC 50 values ranging from ∼40-120 µM. The monounsaturation of LPC-18:1 had no effect when compared to its saturated counterpart LPC-18:0. On the other hand, head group size played a more significant role in blocking fusion as lysophosphatidic acid (LPA)-18:1 failed to fully inhibit fusion. We also show that both Ca 2+ uptake and SNARE-dependent Ca 2+ efflux was sensitive to changes in the acyl chain length and saturation of LPCs, while LPA only affected Ca 2+ efflux. Finally, we tested these LPLs on vacuole acidification by the V-ATPase. This showed that LPC-18:0 could fully inhibit acidification whereas other LPCs had moderate effects. Again, LPA had no effect. Together these data suggest that the effects of LPLs were due to a combination of head group size and acyl chain length leading to a range in degree of positive curvature.
Collapse
|
20
|
Yang C, Li Y, Wu Q, Tang J, Chen M, Zhang B, Li B, Qin Y, Huang G, Zhang Y, Zhi F, Liu K. Unveiling the Pharmacological Role of Human Deubiquitinating Enzymes in Temozolomide Response of Glioblastoma Cells. Cell Biochem Biophys 2024; 82:2183-2193. [PMID: 38809352 DOI: 10.1007/s12013-024-01325-6] [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] [Accepted: 05/17/2024] [Indexed: 05/30/2024]
Abstract
Temozolomide (TMZ) stands as the primary chemotherapeutic drug utilized in clinical glioma treatment, particularly for high-grade glioblastoma (GBM). However, the emergence of TMZ resistance in GBM poses a significant hurdle to its clinical efficacy. Our objective was to elucidate the role of deubiquitinating enzymes (DUBs) in GBM cell resistance to TMZ. We employed the broad-spectrum DUBs inhibitor G5 to investigate the function of DUBs in TMZ cytotoxicity against GBM cells. Eighty-two GBM cell lines with specified DUBs knockout were generated and subjected to CCK-8 assays to assess cell proliferation and TMZ resistance. Furthermore, the association between DUBs and TMZ resistance in GBM cells, along with the modulation of autophagic flux, was examined. The pan-DUBs inhibitor G5 demonstrated the ability to induce cell death and enhance TMZ toxicity in GBM cells. Subsequently, we identified potential DUBs involved in regulating GBM cell proliferation and TMZ resistance. The impact of DUBs knockout on TMZ cytotoxicity was found to be associated with their regulation of TMZ-induced autophagy. In summary, our study provides primary insights into the role of DUBs in GBM cell proliferation and TMZ resistance, and contributes to a deeper understanding of the complex function of DUBs genes underlying TMZ resistance in GBM cells.
Collapse
Affiliation(s)
- Chunguang Yang
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, Guangxi, 530004, China
- Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510630, China
| | - Yifei Li
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, Guangxi, 530004, China
| | - Qifan Wu
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, Guangxi, 530004, China
| | - Jiayi Tang
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, Guangxi, 530004, China
| | - Min Chen
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, Guangxi, 530004, China
| | - Baoyu Zhang
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, China
| | - Bowen Li
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, 213000, China
| | - Yunfei Qin
- Biotherapy Center, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, 510630, China
| | - Guobin Huang
- College of Life and Health Sciences, Guangdong Industry Polytechnic, Guangzhou, Guangdong, 510300, China
| | - Yize Zhang
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, China.
| | - Feng Zhi
- Department of Neurosurgery, The First People's Hospital of Changzhou, Changzhou, Jiangsu, 213000, China.
| | - Kunpeng Liu
- Guangxi Key Laboratory of Special Biomedicine; School of Medicine, Guangxi University, Nanning, Guangxi, 530004, China.
| |
Collapse
|
21
|
Abd El-Mawgoud HK, AboulMagd AM, Nemr MTM, Hemdan MM, Hassaballah AI, Farag PS. Design, synthesis and cytotoxic evaluation of new thieno[2,3-d]pyrimidine analogues as VEGFR-2/AKT dual inhibitors, apoptosis and autophagy inducers. Bioorg Chem 2024; 150:107622. [PMID: 38996545 DOI: 10.1016/j.bioorg.2024.107622] [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: 05/16/2024] [Revised: 06/22/2024] [Accepted: 07/08/2024] [Indexed: 07/14/2024]
Abstract
Novel thieno[2,3-d]pyrimidine analogues were designed, synthesized and evaluated for anti-proliferative activity against HepG-2, PC-3 and MCF-7 cancer cell lines. In addition, WI-38 normal cell line was used to explore the safety of all the tested compounds. Compounds 2 (IC50 = 4.29 µM HePG-2, 10.84 µM MCF-7), 6 (IC50 = 14.86 μM HePG-2, 8.04 μM PC-3 and 12.90 μM MCF-7) and 17 (IC50 = 9.98 μM HePG-2, 33.66 μM PC-3 and 14.62 μM MCF-7) were the most promising candidates on the tested cancer cells with high selective toxicity-sparing normal cells. A further mechanistic evaluation revealed promising kinase inhibitory activity, where compound 2 inhibited VEGFR-2 and AKT at IC50 = 0.161 and 1.06 μM, respectively, Furthermore, derivative 6 inhibited VEGFR-2 and AKT at IC50 = 0.487 and 0.364 μM, respectively, while compound 17 showed IC50 = 0.164 and 0.452 μM, respectively. Moreover, compounds 2, 6 resulted in G1 phase cell cycle arrest while candidate 17 arrest cell cycle at G2/M phase. Similar to the apoptosis results, compound 17 showed the highest autophagic induction among the evaluated derivatives. Finally, docking studies were conducted to assess the binding patterns of these active derivatives. The results showed that the binding patterns inside the active sites of both the VEGFR-2 and AKT-1 (allosteric pocket) crystal structures were identical to the reference ligands.
Collapse
Affiliation(s)
- Heba K Abd El-Mawgoud
- Chemistry Department, Faculty of Women for Arts, Science and Education, Ain Shams University, Heliopolis, 11767 Cairo, Egypt.
| | - Asmaa M AboulMagd
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Nahda University (NUB), Beni-Suef, Egypt.
| | - Mohamed T M Nemr
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr El-Eini Street 11562, Cairo, Egypt
| | - Magdy M Hemdan
- Chemistry Department, Faculty of Science, Ain Shams University, Abbasia, 11566 Cairo, Egypt
| | - Aya I Hassaballah
- Chemistry Department, Faculty of Science, Ain Shams University, Abbasia, 11566 Cairo, Egypt
| | - Paula S Farag
- Chemistry Department, Faculty of Science, Ain Shams University, Abbasia, 11566 Cairo, Egypt
| |
Collapse
|
22
|
Turkoglu B, Mansuroglu B. Investigating the Effects of Chelidonic Acid on Oxidative Stress-Induced Premature Cellular Senescence in Human Skin Fibroblast Cells. Life (Basel) 2024; 14:1070. [PMID: 39337855 PMCID: PMC11433492 DOI: 10.3390/life14091070] [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: 07/30/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
This study investigated the effects of chelidonic acid (CA) on hydrogen peroxide (H2O2) induced cellular senescence in human skin fibroblast cells (BJ). Cellular senescence is a critical mechanism that is linked to age-related diseases and chronic conditions. CA, a γ-pyrone compound known for its broad pharmacological activity, was assessed for its potential to mitigate oxidative stress and alter senescence markers. A stress-induced premature senescence (SIPS) model was designed in BJ fibroblast cells using the oxidative stress agent H2O2. After this treatment, cells were treated with CA, and the potential effect of CA on senescence was evaluated using senescence-related β-galactosidase, 4',6-diamino-2-phenylindole (DAPI), acridine-orange staining (AO), comet assay, molecular docking assays, gene expression, and protein analysis. These results demonstrate that CA effectively reduces senescence markers, including senescence-associated β-galactosidase activity, DNA damage, lysosomal activity, and oxidative stress indicators such as malondialdehyde. Molecular docking revealed CA's potential interactions with critical proteins involved in senescence signalling pathways, suggesting mechanisms by which CA may exert its effects. Gene expression and protein analyses corroborated the observed anti-senescent effects, with CA modulating p16, p21, and pRB1 expressions and reducing oxidative stress markers. In conclusion, CA appeared to have senolytic and senomorphic potential in vitro, which could mitigate and reverse SIPS markers in BJ fibroblasts.
Collapse
Affiliation(s)
| | - Banu Mansuroglu
- Department of Molecular Biology and Genetics, Faculty of Arts and Science, Yildiz Technical University, Istanbul 34220, Turkey;
| |
Collapse
|
23
|
Yu J, Fu Y, Gao J, Zhang Q, Zhang N, Zhang Z, Jiang X, Chen C, Wen Z. Cathepsin C from extracellular histone-induced M1 alveolar macrophages promotes NETosis during lung ischemia-reperfusion injury. Redox Biol 2024; 74:103231. [PMID: 38861835 PMCID: PMC11209641 DOI: 10.1016/j.redox.2024.103231] [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: 04/10/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024] Open
Abstract
Primary graft dysfunction (PGD) is a severe form of acute lung injury resulting from lung ischemia/reperfusion injury (I/R) in lung transplantation (LTx), associated with elevated post-transplant morbidity and mortality rates. Neutrophils infiltrating during reperfusion are identified as pivotal contributors to lung I/R injury by releasing excessive neutrophil extracellular traps (NETs) via NETosis. While alveolar macrophages (AMs) are involved in regulating neutrophil chemotaxis and infiltration, their role in NETosis during lung I/R remains inadequately elucidated. Extracellular histones constitute the main structure of NETs and can activate AMs. In this study, we confirmed the significant involvement of extracellular histone-induced M1 phenotype of AMs (M1-AMs) in driving NETosis during lung I/R. Using secretome analysis, public protein databases, and transwell co-culture models of AMs and neutrophils, we identified Cathepsin C (CTSC) derived from AMs as a major mediator in NETosis. Further elucidating the molecular mechanisms, we found that CTSC induced NETosis through a pathway dependent on NADPH oxidase-mediated production of reactive oxygen species (ROS). CTSC could significantly activate p38 MAPK, resulting in the phosphorylation of the NADPH oxidase subunit p47phox, thereby facilitating the trafficking of cytoplasmic subunits to the cell membrane and activating NADPH oxidase. Moreover, CTSC up-regulated and activated its substrate membrane proteinase 3 (mPR3), resulting in an increased release of NETosis-related inflammatory factors. Inhibiting CTSC revealed great potential in mitigating NETosis-related injury during lung I/R. These findings suggests that CTSC from AMs may be a crucial factor in mediating NETosis during lung I/R, and targeting CTSC inhition may represent a novel intervention for PGD in LTx.
Collapse
Affiliation(s)
- Jing Yu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Department of Anesthesiology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| | - Yu Fu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiameng Gao
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qingqing Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Nan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyuan Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuemei Jiang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Zongmei Wen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
24
|
Park W, Jang H, Kim HS, Park SJ, Lim W, Song G, Park S. Therapeutic efficacy and anti-inflammatory mechanism of baicalein on endometriosis progression in patient-derived cell line and mouse model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155469. [PMID: 38824821 DOI: 10.1016/j.phymed.2024.155469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/22/2024] [Accepted: 02/19/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Baicalein is a flavonoid extracted from the roots of Scutellaria baicalensis G. that has anti-inflammatory and antitumor effects. However, therapeutic mechanisms of baicalein in patients with endometriosis in vivo have yet to be elucidated. As a chronic inflammatory gynecological disease, endometriosis causes pain and infertility, and has no complete treatment to date. Current treatment strategies cause several side effects and have high recurrence rates. PURPOSE This study aimed to identify the in vivo therapeutic effects of baicalein on endometriosis and verify the action mechanisms of baicalein, focusing on regulating inflammation. METHODS In this study, an autologous transplant mouse model and patient-derived immortalized human ovarian endometriotic stromal cells (ihOESCs) were used to investigate the therapeutic activities of baicalein. The mouse model was administered with 40 mg/kg baicalein by oral gavage for 4 weeks, and the treatment outcomes of baicalein-treated mice were compared with vehicle- and dienogest-treated groups. ihOESCs were treated with 0-5 μg/ml baicalein for in vitro studies. RESULTS Baicalein significantly alleviated the progression of endometriosis in mouse models. Baicalein reduced the expression of proinflammatory cytokines in endometriotic lesions and ihOESCs, and cytokine expression and T cell proportions in mouse spleen. in vitro results showed that baicalein increased mitochondrial calcium flux and induced mitochondrial depolarization and ROS generation in ihOESCs. Ultimately, baicalein inactivated the MAPK/PI3K signaling and induced cell death in ihOESCs. CONCLUSION In conclusion, baicalein effectively attenuated the progression of endometriosis through its anti-inflammatory activities. Baicalein can be an alternative or supplemental treatment for endometriosis to ameliorate the side effects of hormonal therapy.
Collapse
Affiliation(s)
- Wonhyoung Park
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Hyewon Jang
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hee Seung Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Soo Jin Park
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea
| | - Whasun Lim
- Department of Biological Sciences, College of Science, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| | - Sunwoo Park
- Department of Plant & Biomaterials Science, Gyeongsang National University, Jinju, 52725, Republic of Korea; Department of GreenBio Science, Gyeongsang National University, Jinju, 52725, Republic of Korea.
| |
Collapse
|
25
|
Han S, Zhuang H, Diao Y, Segal M, Tithi TI, Zhang W, Reeves WH. Altered lipid homeostasis and autophagy precipitate diffuse alveolar hemorrhage in murine lupus. AUTOPHAGY REPORTS 2024; 3:2379193. [PMID: 39871963 PMCID: PMC11772013 DOI: 10.1080/27694127.2024.2379193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 07/01/2024] [Accepted: 07/05/2024] [Indexed: 01/29/2025]
Abstract
Abnormal autophagy regulation is implicated in lupus and other autoimmune diseases. We investigated autophagy in the murine pristane-induced lupus model. Pristane causes monocyte/macrophage-mediated endoplasmic reticulum (ER) stress in lung endothelial cells and diffuse alveolar hemorrhage (DAH) indistinguishable from DAH in lupus patients. Enlarged macrophages with abundant lipid droplets containing neutral lipid and exhibiting increased autophagosome staining were observed in the lung and peritoneal macrophages after pristane treatment. Cellular overload of neutral lipid can lead to selective autophagy (lipophagy) of lipid droplets and transport to lysosomes. The autophagy inducer rapamycin decreased neutral lipid staining but aggravated DAH, while an autophagy inhibitor (3-methyladenine) blocked the onset of DAH. Pristane-induced autophagy in macrophages was confirmed by acridine orange assay and LC3 western blot. Pristane also enlarged lysosomal volume and enhanced cathepsin S, D, and K expression while decreasing lysosomal acid lipase activity. If the capacity to degrade neutral lipid into free cholesterol and fatty acids is overwhelmed, lysosomes enlarge and can release cathepsins into the cytoplasm promoting cell death. Increasing lysosomal cholesterol content by blocking the Niemann-Pick C disease protein NPC1 protects against lysosome-dependent cell death. Treatment with NPC1 inhibitors U18666A or cepharanthine, which stabilize lysosomes, normalized lysosomal volume, reversed ER stress, and prevented DAH in pristane-treated mice. We conclude that pristane disrupts lipid homeostasis, promoting autophagy, lysosomal dysfunction, ER stress, and cell death leading to DAH. NPC1 inhibition reverses these abnormalities, preventing DAH. The findings shed light on the role of autophagy and lysosomal dysfunction in the pathogenesis of lupus.
Collapse
Affiliation(s)
- Shuhong Han
- Division of Rheumatology, Allergy, & Clinical Immunology
| | - Haoyang Zhuang
- Division of Rheumatology, Allergy, & Clinical Immunology
| | - Yanpeng Diao
- Division of Nephrology, Hypertension, and Renal Transplantation
| | - Mark Segal
- Division of Nephrology, Hypertension, and Renal Transplantation
| | - Tanzia Islam Tithi
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL32610
| | - Weizhou Zhang
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL32610
| | - Westley H. Reeves
- Division of Rheumatology, Allergy, & Clinical Immunology
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL32610
| |
Collapse
|
26
|
Kim SM, Kim YH, Han GU, Kim SG, Kim BJ, Moon SH, Shin SH, Ryu BY. Elucidating the mechanisms and mitigation strategies for six-phthalate-induced toxicity in male germ cells. Front Cell Dev Biol 2024; 12:1398176. [PMID: 39050888 PMCID: PMC11266291 DOI: 10.3389/fcell.2024.1398176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 06/18/2024] [Indexed: 07/27/2024] Open
Abstract
Phthalate esters (PAEs) are primary plasticizers and endocrine-disrupting chemicals (EDCs) that are extensively used in numerous everyday consumer products. Although the adverse effects of single PAEs have been studied, our understanding of the effect of multiple phthalate exposure on male germ cell vitality remains limited. Therefore, this study aimed to investigate the collective effects of a mixture of PAEs (MP) comprising diethyl-, bis (2-ethylhexyl)-, dibutyl-, diisononyl-, diisobutyl-, and benzyl butyl-phthalates in the proportions of 35, 21, 15, 15, 8, and 5%, respectively, on differentiated male germ cells using GC-1 spermatogonia (spg) cells. As a mixture, MP substantially hindered GC-1 spg cell proliferation at 3.13 μg/mL, with a half-maximal inhibitory concentration of 16.9 μg/mL. Treatment with 25 μg/mL MP significantly induced reactive oxygen species generation and promoted apoptosis. Furthermore, MP activated autophagy and suppressed phosphorylation of phosphoinositide 3-kinase, protein kinase B, and mammalian target of rapamycin (mTOR). The triple inhibitor combination treatment comprising parthenolide, N-acetylcysteine, and 3-methyladenine effectively reversed MP-induced GC-1 spg cell proliferation inhibition, mitigated apoptosis and autophagy, and restored mTOR phosphorylation. This study is the first to elucidate the mechanism underlying MP-induced male germ cell toxicity and the restoration of male germ cell proliferation mediated by chemical inhibitors. Therefore, it provides valuable insights into the existing literature by proposing a combinatorial toxicity mitigation strategy to counteract male germ cell toxicity induced by various EDCs exposure.
Collapse
Affiliation(s)
- Seok-Man Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, Republic of Korea
| | - Yong-Hee Kim
- AttisLab Inc., Anyang-Si, Gyeonggi-Do, Republic of Korea
| | - Gil Un Han
- Department of Animal Science and Technology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, Republic of Korea
| | - Seul Gi Kim
- Department of Animal Science and Technology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, Republic of Korea
| | - Bang-Jin Kim
- Department of Surgery, Division of Surgical Sciences, Columbia University Irving Medical Center, New York, NY, United States
| | - Sung-Hwan Moon
- Department of Animal Science and Technology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, Republic of Korea
| | - Seung Hee Shin
- Department of Animal Science and Technology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, Republic of Korea
| | - Buom-Yong Ryu
- Department of Animal Science and Technology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, Republic of Korea
| |
Collapse
|
27
|
Kamm M, Hildebrandt F, Titze B, Höink AJ, Vorwerk H, Sievert KD, Groetzner J, Titze U. Ex Vivo Fluorescence Confocal Microscopy for Intraoperative Examinations of Lung Tumors as Alternative to Frozen Sections-A Proof-of-Concept Study. Cancers (Basel) 2024; 16:2221. [PMID: 38927926 PMCID: PMC11202023 DOI: 10.3390/cancers16122221] [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: 05/06/2024] [Revised: 06/09/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Intraoperative frozen sections (FS) are frequently used to establish the diagnosis of lung cancer when preoperative examinations are not conclusive. The downside of FS is its resource-intensive nature and the risk of tissue depletion when small lesions are assessed. Ex vivo fluorescence confocal microscopy (FCM) is a novel microimaging method for loss-free examinations of native materials. We tested its suitability for the intraoperative diagnosis of lung tumors. METHODS Samples from 59 lung resection specimens containing 45 carcinomas were examined in the FCM. The diagnostic performance in the evaluation of malignancy and histological typing of lung tumors was evaluated in comparison with FS and the final diagnosis. RESULTS A total of 44/45 (98%) carcinomas were correctly identified as malignant in the FCM. A total of 33/44 (75%) carcinomas were correctly subtyped, which was comparable with the results of FS and conventional histology. Our tests documented the excellent visualization of cytological features of normal tissues and tumors. Compared to FS, FCM was technically less demanding and less personnel intensive. CONCLUSIONS The ex vivo FCM is a fast, effective, and safe method for diagnosing and subtyping lung cancer and is, therefore, a promising alternative to FS. The method preserves the tissue without loss for subsequent examinations, which is an advantage in the diagnosis of small tumors and for biobanking.
Collapse
Affiliation(s)
- Max Kamm
- Department of Pathology, Medical School and University Medical Center OWL, Klinikum Lippe, Lung Cancer Center Lippe, Bielefeld University, 32756 Detmold, Germany; (M.K.); (F.H.); (B.T.)
| | - Felix Hildebrandt
- Department of Pathology, Medical School and University Medical Center OWL, Klinikum Lippe, Lung Cancer Center Lippe, Bielefeld University, 32756 Detmold, Germany; (M.K.); (F.H.); (B.T.)
| | - Barbara Titze
- Department of Pathology, Medical School and University Medical Center OWL, Klinikum Lippe, Lung Cancer Center Lippe, Bielefeld University, 32756 Detmold, Germany; (M.K.); (F.H.); (B.T.)
| | - Anna Janina Höink
- Department of Diagnostic and Interventional Radiology, Medical School and University Medical Center OWL, Klinikum Lippe, Lung Cancer Center Lippe, Bielefeld University, 32756 Detmold, Germany;
| | - Hagen Vorwerk
- Department of Pneumology, Respiratory and Sleep Medicine, Klinikum Lippe Lemgo, Lung Cancer Center Lippe, 32657 Lemgo, Germany;
| | - Karl-Dietrich Sievert
- Department of Urology, Medical School and University Medical Center OWL, Klinikum Lippe, Bielefeld University, 32756 Detmold, Germany;
| | - Jan Groetzner
- Department of Thoracic Surgery, Klinikum Lippe Lemgo, Lung Cancer Center Lippe, 32657 Lemgo, Germany;
| | - Ulf Titze
- Department of Pathology, Medical School and University Medical Center OWL, Klinikum Lippe, Lung Cancer Center Lippe, Bielefeld University, 32756 Detmold, Germany; (M.K.); (F.H.); (B.T.)
| |
Collapse
|
28
|
Shin JS, Jang Y, Kim DS, Jung E, Lee MK, Kim B, Ahn S, Shin Y, Jang SS, Yun CS, Yoo J, Lim YC, Han SB, Kim M. Inhibition of endocytic uptake of severe acute respiratory syndrome coronavirus 2 and endo-lysosomal acidification by diphenoxylate. Antimicrob Agents Chemother 2024; 68:e0034124. [PMID: 38742905 PMCID: PMC11620506 DOI: 10.1128/aac.00341-24] [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: 03/04/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024] Open
Abstract
Cell culture-based screening of a chemical library identified diphenoxylate as an antiviral agent against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The observed 50% effective concentrations ranged between 1.4 and 4.9 µM against the original wild-type strain and its variants. Time-of-addition experiments indicated that diphenoxylate is an entry blocker targeting a host factor involved in viral infection. Fluorescence microscopic analysis visualized that diphenoxylate prevented SARS-CoV-2 particles from penetrating the cell membrane and also impaired endo-lysosomal acidification. Diphenoxylate exhibited a synergistic inhibitory effect on SARS-CoV-2 infection in human lung epithelial Calu-3 cells when combined with a transmembrane serine protease 2 (TMPRSS2) inhibitor, nafamostat. This synergy suggested that efficient antiviral activity is achieved by blocking both TMPRSS2-mediated early and endosome-mediated late SARS-CoV-2 entry pathways. The antiviral efficacy of diphenoxylate against SARS-CoV-2 was reproducible in a human tonsil organoids system. In a transgenic mouse model expressing the obligate SARS-CoV-2 receptor, human angiotensin-converting enzyme 2, intranasal administration of diphenoxylate (10 mg/kg/day) significantly reduced the viral RNA copy number in the lungs by 70% on day 3. This study underscores that diphenoxylate represents a promising core scaffold, warranting further exploration for chemical modifications aimed at developing a new class of clinically effective antiviral drugs against SARS-CoV-2.
Collapse
Affiliation(s)
- Jin Soo Shin
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Yejin Jang
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Dong-Su Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Eunhye Jung
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Myoung Kyu Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Byungil Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Sunjoo Ahn
- Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Yeonju Shin
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Su San Jang
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Chang Soo Yun
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| | - Jongman Yoo
- CHA Organoid Research Center, CHA University, Seongnam, Gyeonggi-do, Republic of Korea
| | - Young Chang Lim
- Department of Otorhinolaryngology-Head and Neck Surgery, The Research Institute, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Soo Bong Han
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
- Medicinal Chemistry and Pharmacology, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Meehyein Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, Republic of Korea
| |
Collapse
|
29
|
Yaya-Candela AP, Ravagnani FG, Dietrich N, Sousa R, Baptista MS. Specific photodamage on HT-29 cancer cells leads to endolysosomal failure and autophagy blockage by cathepsin depletion. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 255:112919. [PMID: 38677261 DOI: 10.1016/j.jphotobiol.2024.112919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
Endolysosomes perform a wide range of cellular functions, including nutrient sensing, macromolecule digestion and recycling, as well as plasma membrane repair. Because of their high activity in cancerous cells, endolysosomes are attractive targets for the development of novel cancer treatments. Light-activated compounds termed photosensitizers (PS) can catalyze the oxidation of specific biomolecules and intracellular organelles. To selectively damage endosomes and lysosomes, HT-29 colorectal cancer cells were incubated with nanomolar concentrations of meso-tetraphenylporphine disulfonate (TPPS2a), an amphiphilic PS taken up via endocytosis and activated by green light (522 nm, 2.1 J.cm-1). Several cellular responses were characterized by a combination of immunofluorescence and immunoblotting assays. We showed that TPPS2a photosensitization blocked autophagic flux without extensive endolysosomal membrane rupture. Nevertheless, there was a severe functional failure of endolysosomes due to a decrease in CTSD (cathepsin D, 55%) and CTSB (cathepsin B, 52%) maturation. PSAP (prosaposin) processing (into saposins) was also considerably impaired, a fact that could be detrimental to glycosphingolipid homeostasis. Therefore, photosensitization of HT-29 cells previously incubated with a low concentration of TPPS2a promotes endolysosomal dysfunction, an effect that can be used to improve cancer therapies.
Collapse
Affiliation(s)
| | | | - Natasha Dietrich
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Rafaela Sousa
- Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
30
|
Madrid Mendoza MF, Almeida Mota J, de Cassia Evangelista de Oliveira F, Cavalcanti BC, Fabio Turco J, Reyes Torres Y, Ferreira PMP, Barros-Nepomuceno FWA, Rocha DD, Pessoa C, de Moraes Filho MO. Ethanolic extract from leaves of tithonia diversifolia induces apoptosis in HCT-116 cells through oxidative stress. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2024; 87:275-293. [PMID: 38285019 DOI: 10.1080/15287394.2024.2308256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2024]
Abstract
Tithonia diversifolia is a perennial bushy plant found in South America with significant ethnopharmacological importance as an antimalarial, antidiabetic, antibacterial, and anticancer agent. The aim of the present study was to determine the cytotoxicity of the ethanolic extract from leaves of T. diversifolia (TdE) on human cancer cell lines (HCT-116, SNB-19, NCIH-460 and MCF-7), as well as the mechanism of action involved in cell death and cellular modulation of oxidative stress. The TdE exhibited significant activity with IC50 values ranging from 7.12 to 38.41 μg/ml, with HCT-116 being the most sensitive cell line. Subsequent experiments were conducted with HCT-116 cell line. TdE decreased the number of viable cells, followed by induction of apoptotic events, increase in mitochondrial membrane permeabilization, and enhanced G2/M phase of the cell cycle. Pro-oxidative effects including elevated acidic vesicular organelle formation, lipid peroxidation, and nitric oxide by-products, as well as reduced levels of intracellular glutathione and reactive oxygen species production were also observed following incubation with TdE, which may lead to DNA damage followed by apoptotic cell death. These results demonstrate the potential of TdE ethanolic leaf extraction for biological activity and enhance the importance of continuing to study natural sources of plants for the development of anticancer agents.
Collapse
Affiliation(s)
| | - Jessica Almeida Mota
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | | | | | - João Fabio Turco
- Department of Chemistry, Midwestern State University of Guarapuava, Guarapuava, Brazil
| | - Yohandra Reyes Torres
- Department of Chemistry, Midwestern State University of Guarapuava, Guarapuava, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| | | | - Danilo Damasceno Rocha
- Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Brazil
| | - Claudia Pessoa
- Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | | |
Collapse
|
31
|
Venz R, Goyala A, Soto-Gamez A, Yenice T, Demaria M, Ewald CY. In-vivo screening implicates endoribonuclease Regnase-1 in modulating senescence-associated lysosomal changes. GeroScience 2024; 46:1499-1514. [PMID: 37644339 PMCID: PMC10828269 DOI: 10.1007/s11357-023-00909-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 08/07/2023] [Indexed: 08/31/2023] Open
Abstract
Accumulation of senescent cells accelerates aging and age-related diseases, whereas preventing this accumulation extends the lifespan in mice. A characteristic of senescent cells is increased staining with β-galactosidase (β-gal) ex vivo. Here, we describe a progressive accumulation of β-gal staining in the model organism C. elegans during aging. We show that distinct pharmacological and genetic interventions targeting the mitochondria and the mTORC1 to the nuclear core complex axis, the non-canonical apoptotic, and lysosomal-autophagy pathways slow the age-dependent accumulation of β-gal. We identify a novel gene, rege-1/Regnase-1/ZC3H12A/MCPIP1, modulating β-gal staining via the transcription factor ets-4/SPDEF. We demonstrate that knocking down Regnase-1 in human cell culture prevents senescence-associated β-gal accumulation. Our data provide a screening pipeline to identify genes and drugs modulating senescence-associated lysosomal phenotypes.
Collapse
Affiliation(s)
- Richard Venz
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, CH-8603, Schwerzenbach, Switzerland
| | - Anita Goyala
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, CH-8603, Schwerzenbach, Switzerland
| | - Abel Soto-Gamez
- European Institute for the Biology of Aging (ERIBA)/University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Tugce Yenice
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, CH-8603, Schwerzenbach, Switzerland
| | - Marco Demaria
- European Institute for the Biology of Aging (ERIBA)/University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Collin Y Ewald
- Laboratory of Extracellular Matrix Regeneration, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, CH-8603, Schwerzenbach, Switzerland.
| |
Collapse
|
32
|
Shen Y, Li X, Wang H, Wang Y, Tao L, Wang P, Zhang H. Bisphenol A induced neuronal apoptosis and enhanced autophagy in vitro through Nrf2/HO-1 and Akt/mTOR pathways. Toxicology 2023; 500:153678. [PMID: 38006930 DOI: 10.1016/j.tox.2023.153678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 11/10/2023] [Accepted: 11/18/2023] [Indexed: 11/27/2023]
Abstract
Bisphenol A (BPA) was traditionally used in epoxy resins and polycarbonate plastics, but it was found to be harmful to human health due to its endocrine-disrupting effects. It can affect various biological functions of human beings and interfere with brain development. However, the neurotoxic mechanisms of BPA on brain development and associated neurodegeneration remain poorly understood. Here, we reported that BPA (100, 250, 500 μM) inhibited cell viability of neural cells PC12, SH-SY5Y and caused dose-dependent cell death. In addition, BPA exposure increased intracellular reactive oxygen species (ROS) and mitochondrial ROS (mtROS) levels, decreased mitochondrial membrane potential, reduced the expression of cytochrome c oxidase IV (COX4), downregulated Bcl-2, and initiated apoptosis. Moreover, BPA treatment resulted in the accumulation of intracellular acidic vacuoles and increased the autophagy marker LC3 II to LC3 I ratio. Furthermore, BPA exposure inhibited Nrf2/ HO-1 and AKT/mTOR pathways and mediated cellular oxidative stress, apoptosis, and excessive autophagy, leading to neuronal degeneration. The interactions between oxidative stress, autophagy, and apoptosis during BPA-induced neurotoxicity remain unclear and require further in vivo confirmation.
Collapse
Affiliation(s)
- Yue Shen
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Xinying Li
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Hongyan Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yicheng Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Liqing Tao
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China; School of Life Sciences, Shaoxing University, Shaoxing, Zhejiang, China
| | - Pingping Wang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China
| | - Heng Zhang
- Neurodegeneration and Neuroregeneration Laboratory, Department of Basic Medicine, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China; School of Life Sciences, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
33
|
Spano A, Sciola L. Polyploid cell dynamics and death before and after PEG-treatment of a NIH/3T3 derived culture: vinblastine effects on the regulation of cell subpopulations heterogeneity. Cell Div 2023; 18:18. [PMID: 37904245 PMCID: PMC10614354 DOI: 10.1186/s13008-023-00100-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/14/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND Neoplastic subpopulations can include polyploid cells that can be involved in tumor evolution and recurrence. Their origin can be traced back to the tumor microenvironment or chemotherapeutic treatment, which can alter cell division or favor cell fusion, generating multinucleated cells. Their progeny, frequently genetically unstable, can result in new aggressive and more resistant to chemotherapy subpopulations. In our work, we used NIHs cells, previously derived from the NIH/3T3 line after serum deprivation, that induced a polyploidization increase with the appearance of cells with DNA content ranging from 4 to 24c. This study aimed to analyze the cellular dynamics of NIHs culture subpopulations before and after treatment with the fusogenic agent polyethylene glycol (PEG), which allowed us to obtain new giant polyploid cells. Successively, PEG-untreated and PEG-treated cultures were incubated with the antimicrotubular poison vinblastine. The dynamics of appearance, decrease and loss of cell subpopulations were evaluated by correlating cell DNA content to mono-multinuclearity resulting from cell fusion and division process alteration and to the peculiarities of cell death events. RESULTS DNA microfluorimetry and morphological techniques (phase contrast, fluorescence and TEM microscopies) indicated that PEG treatment induced a 4-24c cell increase and the appearance of new giant elements (64-140c DNA content). Ultrastructural analysis and autophagosomal-lysosomal compartment fluorochromization, which allowed us to correlate cytoplasmic changes to death events, indicated that cell depletion occurred through distinct mechanisms: apoptotic death involved 2c, 4c and 8c cells, while autophagic-like death involved intermediate 12-24c cells, showing nuclear (lobulation/micronucleation) and autophagic cytoplasm alterations. Death, spontaneously occurring, especially in intermediate-sized cells, was increased after vinblastine treatment. No evident cell loss by death events was detected in the 64-140c range. CONCLUSIONS PEG-treated NIHs cultures can represent a model of heterogeneous subpopulations originating from cell fusion and division process anomalies. Altogether, our results suggest that the different cell dynamics of NIHs subpopulations can affect the variability of responses to stimuli able to induce cell degeneration and death. Apoptptic, autophagic or hybrid forms of cell death can also depend on the DNA content and ability to progress through the cell cycle, which may influence the persistence and fate of polyploid cell descendants, also concerning chemotherapeutic agent action.
Collapse
Affiliation(s)
- Alessandra Spano
- Department of Biomedical Sciences, Sassari - University of Sassari, Via Muroni 25, 07100, Sassari, Italy
| | - Luigi Sciola
- Department of Biomedical Sciences, Sassari - University of Sassari, Via Muroni 25, 07100, Sassari, Italy.
| |
Collapse
|
34
|
Omata K, Nomura I, Hirata A, Yonezuka Y, Muto H, Kuriki R, Jimbo K, Ogasa K, Kato T. Isolation and evaluation of erythroid progenitors in the livers of larval, froglet, and adult Xenopus tropicalis. Biol Open 2023; 12:bio059862. [PMID: 37421150 PMCID: PMC10399205 DOI: 10.1242/bio.059862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 06/28/2023] [Indexed: 07/09/2023] Open
Abstract
Xenopus liver maintains erythropoietic activity from the larval to the adult stage. During metamorphosis, thyroid hormone mediates apoptosis of larval-type erythroid progenitors and proliferation of adult-type erythroid progenitors, and a globin switch occurs during this time. In addition, the whole-body mass and the liver also change; however, whether there is a change in the absolute number of erythroid progenitors is unclear. To isolate and evaluate erythroid progenitors in the Xenopus liver, we developed monoclonal ER9 antibodies against the erythropoietin receptor (EPOR) of Xenopus. ER9 recognized erythrocytes, but not white blood cells or thrombocytes. The specificity of ER9 for EPOR manifested as its inhibitory effect on the proliferation of a Xenopus EPOR-expressing cell line. Furthermore, ER9 recognition was consistent with epor gene expression. ER9 staining with Acridine orange (AO) allowed erythrocyte fractionation through fluorescence-activated cell sorting. The ER9+ and AO-red (AOr)high fractions were highly enriched in erythroid progenitors and primarily localized to the liver. The method developed using ER9 and AO was also applied to larvae and froglets with different progenitor populations from adult frogs. The liver to body weight and the number of ER9+ AOrhigh cells per unit body weight were significantly higher in adults than in larvae and froglets, and the number of ER9+ AOrhigh cells per unit liver weight was the highest in froglets. Collectively, our results show increased erythropoiesis in the froglet liver and demonstrate growth-dependent changes in erythropoiesis patterns in specific organs of Xenopus.
Collapse
Affiliation(s)
- Kazuki Omata
- Department of Biology, School of Education, Waseda University, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
- Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
| | - Ikki Nomura
- Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
| | - Akito Hirata
- Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
| | - Yuka Yonezuka
- Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
| | - Hiroshi Muto
- Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
| | - Ryo Kuriki
- Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
| | - Kirin Jimbo
- Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
| | - Koujin Ogasa
- Department of Biology, School of Education, Waseda University, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
| | - Takashi Kato
- Department of Biology, School of Education, Waseda University, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
- Graduate School of Advanced Science and Engineering, Waseda University, 2-2 Wakamatsu, Shinjuku, Tokyo 162-8480, Japan
| |
Collapse
|
35
|
Kim SH, Cho YS, Kim Y, Park J, Yoo SM, Gwak J, Kim Y, Gwon Y, Kam TI, Jung YK. Endolysosomal impairment by binding of amyloid beta or MAPT/Tau to V-ATPase and rescue via the HYAL-CD44 axis in Alzheimer disease. Autophagy 2023; 19:2318-2337. [PMID: 36843263 PMCID: PMC10351450 DOI: 10.1080/15548627.2023.2181614] [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: 04/14/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/28/2023] Open
Abstract
Impaired activities and abnormally enlarged structures of endolysosomes are frequently observed in Alzheimer disease (AD) brains. However, little is known about whether and how endolysosomal dysregulation is triggered and associated with AD. Here, we show that vacuolar ATPase (V-ATPase) is a hub that mediates proteopathy of oligomeric amyloid beta (Aβ) and hyperphosphorylated MAPT/Tau (p-MAPT/Tau). Endolysosomal integrity was largely destroyed in Aβ-overloaded or p-MAPT/Tau-positive neurons in culture and AD brains, which was a necessary step for triggering neurotoxicity, and treatments with acidic nanoparticles or endocytosis inhibitors rescued the endolysosomal impairment and neurotoxicity. Interestingly, we found that the lumenal ATP6V0C and cytosolic ATP6V1B2 subunits of the V-ATPase complex bound to the internalized Aβ and cytosolic PHF-1-reactive MAPT/Tau, respectively. Their interactions disrupted V-ATPase activity and accompanying endolysosomal activity in vitro and induced neurodegeneration. Using a genome-wide functional screen, we isolated a suppressor, HYAL (hyaluronidase), which reversed the endolysosomal dysfunction and proteopathy and alleviated the memory impairment in 3xTg-AD mice. Further, we found that its metabolite hyaluronic acid (HA) and HA receptor CD44 attenuated neurotoxicity in affected neurons via V-ATPase. We propose that endolysosomal V-ATPase is a bona fide proteotoxic receptor that binds to pathogenic proteins and deteriorates endolysosomal function in AD, leading to neurodegeneration in proteopathy.Abbreviations: AAV, adeno-associated virus; Aβ, amyloid beta; AD, Alzheimer disease; APP, amyloid beta precursor protein; ATP6V0C, ATPase H+ transporting V0 subunit c; ATP6V1A, ATPase H+ transporting V1 subunit A; ATP6V1B2, ATPase H+ transporting V1 subunit B2; CD44.Fc, CD44-mouse immunoglobulin Fc fusion construct; Co-IP, co-immunoprecipitation; CTSD, cathepsin D; HA, hyaluronic acid; HMWHA, high-molecular-weight hyaluronic acid; HYAL, hyaluronidase; i.c.v, intracerebroventricular; LMWHA, low-molecular-weight hyaluronic acid; NPs, nanoparticles; p-MAPT/Tau, hyperphosphorylated microtubule associated protein tau; PI3K, phosphoinositide 3-kinase; V-ATPase, vacuolar-type H+-translocating ATPase; WT, wild-type.
Collapse
Affiliation(s)
- Seo-Hyun Kim
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Young-Sin Cho
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Youbin Kim
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul, Korea
| | - Jisu Park
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Seung-Min Yoo
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Jimin Gwak
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Youngwon Kim
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Youngdae Gwon
- School of Medicine, Sungkyunkwan University, Suwon, Korea
| | - Tae-in Kam
- Department of Neurology and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yong-Keun Jung
- School of Biological Sciences, Seoul National University, Seoul, Korea
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul, Korea
| |
Collapse
|
36
|
Montiel T, Gómora-García JC, Gerónimo-Olvera C, Heras-Romero Y, Bernal-Vicente BN, Pérez-Martínez X, Tovar-Y-Romo LB, Massieu L. Modulation of the autophagy-lysosomal pathway and endoplasmic reticulum stress by ketone bodies in experimental models of stroke. J Neurochem 2023; 166:87-106. [PMID: 37328918 DOI: 10.1111/jnc.15852] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 05/13/2023] [Accepted: 05/17/2023] [Indexed: 06/18/2023]
Abstract
Ischemic stroke is a leading cause of disability worldwide. There is no simple treatment to alleviate ischemic brain injury, as thrombolytic therapy is applicable within a narrow time window. During the last years, the ketogenic diet (KD) and the exogenous administration of the ketone body β-hydroxybutyrate (BHB) have been proposed as therapeutic tools for acute neurological disorders and both can reduce ischemic brain injury. However, the mechanisms involved are not completely clear. We have previously shown that the D enantiomer of BHB stimulates the autophagic flux in cultured neurons exposed to glucose deprivation (GD) and in the brain of hypoglycemic rats. Here, we have investigated the effect of the systemic administration of D-BHB, followed by its continuous infusion after middle cerebral artery occlusion (MCAO), on the autophagy-lysosomal pathway and the activation of the unfolded protein response (UPR). Results show for the first time that the protective effect of BHB against MCAO injury is enantiomer selective as only D-BHB, the physiologic enantiomer of BHB, significantly reduced brain injury. D-BHB treatment prevented the cleavage of the lysosomal membrane protein LAMP2 and stimulated the autophagic flux in the ischemic core and the penumbra. In addition, D-BHB notably reduced the activation of the PERK/eIF2α/ATF4 pathway of the UPR and inhibited IRE1α phosphorylation. L-BHB showed no significant effect relative to ischemic animals. In cortical cultures under GD, D-BHB prevented LAMP2 cleavage and decreased lysosomal number. It also abated the activation of the PERK/eIF2α/ATF4 pathway, partially sustained protein synthesis, and reduced pIRE1α. In contrast, L-BHB showed no significant effects. Results suggest that protection elicited by D-BHB treatment post-ischemia prevents lysosomal rupture allowing functional autophagy, preventing the loss of proteostasis and UPR activation.
Collapse
Affiliation(s)
- Teresa Montiel
- Departamento de Neuropatología Molecular, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Juan Carlos Gómora-García
- Departamento de Neuropatología Molecular, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Cristian Gerónimo-Olvera
- Departamento de Neuropatología Molecular, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Yessica Heras-Romero
- Departamento de Psicobiología y Neurociencias, División de Estudios de Posgrado e Investigación, Facultad de Psicología, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Berenice N Bernal-Vicente
- Departamento de Neuropatología Molecular, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Xochitl Pérez-Martínez
- Departamento de Genética Molecular, División de Investigación Básica, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Luis B Tovar-Y-Romo
- Departamento de Neuropatología Molecular, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Lourdes Massieu
- Departamento de Neuropatología Molecular, División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
37
|
Urade R, Chou CK, Chou HL, Chen BH, Wang TN, Tsai EM, Hung CT, Wu SJ, Chiu CC. Phthalate derivative DEHP disturbs the antiproliferative effect of camptothecin in human lung cancer cells by attenuating DNA damage and activating Akt/NF-κB signaling pathway. ENVIRONMENTAL TOXICOLOGY 2023; 38:332-342. [PMID: 36394428 DOI: 10.1002/tox.23686] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 06/16/2023]
Abstract
Plasticizers/phthalates play a facilitating role in the development of cancer and help the tumor to grow and metastasize. Camptothecin (CPT) and its derivatives are known to have anticancer properties of inhibiting cell growth, promoting cell apoptosis, and increasing autophagy. Therefore, in this study, we investigated whether the presence of di(2-ethylhexyl) phthalate (DEHP) could hinder apoptosis and autophagy caused by CPT in non-small cell lung cancer (NSCLC) cells. We found that DEHP interferes with CPT-induced apoptosis and autophagy and increases the prosurvival pathway by reducing the DNA damage marker γ-H2AX and activating the Akt and NF-κB pathways. Furthermore, we also confirmed that combining DEHP with 3-MA has additive effects in inhibiting autophagy and apoptosis in NSCLC cells. Taken together, our findings show that DEHP could affect CPT-induced anticancer treatment and provide evidence to show that DEHP induces chemoresistance in CPT-based chemotherapy.
Collapse
Affiliation(s)
- Ritesh Urade
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chon-Kit Chou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, People's Republic of China
| | - Han-Lin Chou
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bing-Hung Chen
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsu-Nai Wang
- Department of Public Health, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Eing-Mei Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- The Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Tzu Hung
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shyh-Jong Wu
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Chih Chiu
- Department of Biological Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
38
|
Nath S, Patra D, Nag A, Kundu R. Sesquiterpenoid-rich Java Ginger rhizome extract prompts autophagic cell death in cervical cancer cell SiHa mainly by modulating cellular redox homeostasis. 3 Biotech 2023; 13:8. [PMID: 36532858 PMCID: PMC9751246 DOI: 10.1007/s13205-022-03415-9] [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: 08/23/2022] [Accepted: 11/26/2022] [Indexed: 12/23/2022] Open
Abstract
Java Ginger or Curcuma zanthorrhiza Roxb. has long gained focus among tribal people of Java, for its medicinal properties mainly against gynaecological challenges. The present study aims to identify the most potent phytocompound present in the extract and determine primary mode of action accountable for cytotoxic activity of Curcuma zanthorrhiza rhizome extract against HPV16-positive SiHa cervical cancer cells. The phytochemically-rich extract of rhizome (CZM) was capable to inhibit proliferation of target cells in a dose-dependent manner with an IC50 of 150 μg/ml. Dysregulation of intercellular antioxidant defence system resulted to surges in ROS and RNS level, increased calcium concentration and compromised mitochondrial membrane potential. Nucleus got affected, cell cycle dynamics got impaired while clonogenicity and migration ability diminished. Expression of viral oncogenes E7 and E6 decreased significantly. Accumulation of toxic cell metabolite and decrease in level of essential ones continued. Finally, alteration in PI3K/AKT/mTOR signalling route was followed by onset of autophagic cell death concomitant with the upregulated expression of Beclin1, Atg5-12 and LC3II. Curcumin and a novel crystal as well as few phyto-fractions were isolated by column chromatography. Of these, curcumin was found to be most potent in inducing cytotoxicity in SiHa while two other fractions also showed significant activity. Thus, CZM acted against SiHa cells by inducing autophagy that commences in compliance to the changes in PI3K/AKT/mTOR pathway mainly in response to oxidative stress. To the best of our knowledge this is the first report of Curcuma zanthorrhiza Roxb. inducing autophagy. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-022-03415-9.
Collapse
Affiliation(s)
- Sonali Nath
- Centre of Advanced Study, Department of Botany, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019 India
| | - Debashis Patra
- Department of Chemistry, Taki Government College, Taki, 743426 West Bengal India
| | - Anish Nag
- Department of Life Sciences, Christ University, Bangalore, 560029 Karnataka India
| | - Rita Kundu
- Centre of Advanced Study, Department of Botany, University of Calcutta, 35, Ballygunge Circular Road, Kolkata, 700019 India
| |
Collapse
|
39
|
Luo J, Jin W, Jin M, Pan W, Gao S, Zhao X, Lai X, Sun L, Piao C. Jiedutongluotiaogan formula restores pancreatic function by suppressing excessive autophagy and endoplasmic reticulum stress. PHARMACEUTICAL BIOLOGY 2022; 60:1542-1555. [PMID: 35944284 PMCID: PMC9367665 DOI: 10.1080/13880209.2022.2107019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/22/2022] [Accepted: 07/25/2022] [Indexed: 06/15/2023]
Abstract
CONTEXT Jiedutongluotiaogan formula (JTTF), a traditional Chinese medicine (TCM), could promote islet function. However, the potential effect of JTTF on endoplasmic reticulum stress (ERS) and autophagy have not been reported. OBJECTIVE This study explores the potential effect of JTTF on ERS and autophagy in the pancreas. MATERIALS AND METHODS The Zucker diabetic fatty (ZDF) rats were randomised into five groups, control, model, JTTF (1, 3, 5 g/kg/day for 12 weeks). LPS induced pancreatic β-cells were treated with JTTF (50, 100, 200 μg/mL). LPS was used to induce pancreatic β-cell injury, with cell viability and insulin secretion evaluated using MTT, glucose-stimulated insulin secretion (GSIS) assays, and PCR. Intracellular Ca2+ concentration was measured using flow cytometry, while ERS and autophagy levels were monitored via Western blotting and/or immunostaining. RESULTS Compared with the model group, body weight, FGB, HbA1c, IPGTT, FINs, and HOMA-IR in JTTF treatment groups were significantly reduced. In islets cells treated with JTTF, the pancreatic islet cells in the JTTF group were increased, lipid droplets were reduced, and there was a decrease in Ca2+ (16.67%). After JTTF intervention, PERK, p-PERK, IRE1α, p- IRE1α, ATF6, eIF2α, GRP78, p-ULK1, LC3 and p62 expression decreased, whereas Beclin1and p-mTOR expression increased. In addition, the expression of proteins related to apoptosis in the JTTF groups were lower than those in the control group. DISCUSSION AND CONCLUSIONS JTTF may alleviate pancreatic β-cell injury by inhibiting ER stress and excessive autophagy in diabetic rats. This provides a new direction for treating diabetes and restoring pancreatic dysfunction by TCM.
Collapse
Affiliation(s)
- Jinli Luo
- Institution of Shenzhen Hospital, Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Wenqi Jin
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Meiying Jin
- The Third Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Weiwei Pan
- School of Clinical Medicine, Changchun Medical College, Changchun, China
| | - Shengnan Gao
- Institution of Shenzhen Hospital, Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Xiaohua Zhao
- Institution of Shenzhen Hospital, Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Xingrong Lai
- Institution of Shenzhen Hospital, Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Chunli Piao
- Institution of Shenzhen Hospital, Guangzhou University of Chinese Medicine (Futian), Shenzhen, China
| |
Collapse
|
40
|
Zhou J, Tan Y, Hu L, Fu J, Li D, Chen J, Long Y. Inhibition of HSPA8 by rifampicin contributes to ferroptosis via enhancing autophagy. Liver Int 2022; 42:2889-2899. [PMID: 36254713 DOI: 10.1111/liv.15459] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 08/17/2022] [Accepted: 08/24/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIM Rifampicin is the most common pathogenic factor in anti-tuberculosis drug-induced liver injury (AT-DILI), the mechanisms that it promotes hepatocyte damage in AT-DILI are not yet to be thoroughly elucidated. In this study, we investigated the potential molecular mechanisms for ferroptosis involving rifampicin hepatotoxicity. METHODS Animal and cell injury models of rifampicin were constructed, and the toxicity of rifampicin was assessed by physicochemical staining and cell viability assay. Next, flow cytometry was employed to detect changes in ferroptosis-related markers, and Western blotting was used to detect protein expression. Then, the important role of autophagy and ferroptosis was verified with small molecule compound intervention. RESULTS We found that ferritinophagy-induced ferroptosis participates in the toxicity of rifampicin, and the mechanism is that rifampicin precisely activates high-throughput autophagy, which leads to the massive degradation of ferritin and the increase of free iron. Moreover, rifampicin exhibited conspicuous inhibition of Human 71 kDa heat shock cognate protein (HSPA8) that is intimately associated with Microtubule-associated protein light chain 3 isoform B (LC3B) expression, in turn, HSPA8 inducer attenuated intracellular autophagy flux. Of note, inducing HSPA8 or inhibition of autophagy and ferroptosis considerably relieved the hepatotoxicity of rifampicin in mouse model. CONCLUSIONS The present study highlights the crucial roles of the HSPA8 and autophagy in ferroptotic cell death driving by rifampicin, particularly illumines multiple promising regulatory nodes for therapeutic interventions in diseases involving AT-DILI.
Collapse
Affiliation(s)
- Juan Zhou
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Yingzheng Tan
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Lingli Hu
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Jingli Fu
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Dan Li
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| | - Jun Chen
- Department of Liver Diseases, Third Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yunzhu Long
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, Hunan, China
| |
Collapse
|
41
|
Li X, Wang ZG, Zhu H, Wen HP, Ning D, Liu HY, Pang DW, Liu SL. Inducing Autophagy and Blocking Autophagic Flux via a Virus-Mimicking Nanodrug for Cancer Therapy. NANO LETTERS 2022; 22:9163-9173. [PMID: 36374537 DOI: 10.1021/acs.nanolett.2c04091] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Maximizing the therapeutic capacity of drugs by allowing them to escape lysosomal degradation is a long-term challenge for nanodrug delivery. Japanese encephalitis virus (JEV) has evolved the ability to escape the endosomal region to avoid degradation of internal genetic material by lysosomes and further induce upregulation of cellular autophagy for the purpose of their mass reproduction. In this work, to exploit the lysosome escape and autophagy-inducing properties of JEV for cancer therapy, we constructed a virus-mimicking nanodrug consisting of anti-PDL1 antibody-decorated JEV-mimicking virosome encapsulated with a clinically available autophagy inhibitor, hydroxychloroquine (HCQ). Our study indicated that the nanodrug can upregulate the autophagy level and inhibit the autophagic flux, thereby inducing the apoptosis of tumor cells, and further activating the immune response, which can greatly improve the antitumor and tumor metastasis suppression effects and provide a potential therapeutic strategy for tumor treatment.
Collapse
Affiliation(s)
- Xiao Li
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, P. R. China
| | - Zhi-Gang Wang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, P. R. China
| | - Han Zhu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, P. R. China
| | - Hui-Ping Wen
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, P. R. China
| | - Di Ning
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, P. R. China
| | - Hao-Yang Liu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, P. R. China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, P. R. China
| | - Shu-Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, P. R. China
| |
Collapse
|
42
|
Enhancement of Immunosuppressive Activity of Mesenchymal Stromal Cells by Platelet-Derived Factors is Accompanied by Apoptotic Priming. Stem Cell Rev Rep 2022; 19:713-733. [PMID: 36417151 PMCID: PMC10070232 DOI: 10.1007/s12015-022-10471-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2022] [Indexed: 11/24/2022]
Abstract
Abstract
The pro-inflammatory phase of bone healing, initiated by platelet activation and eventually hematoma formation, impacts bone marrow mesenchymal stromal cells (MSCs) in unknown ways. Here, we created platelet-rich plasma (PRP) hydrogels to study how platelet-derived factors modulate functional properties of encapsulated MSCs in comparison to a non-inflammatory fibrin (FBR) hydrogel environment. MSCs were isolated from human bone marrow, while PRP was collected from pooled apheresis thrombocyte concentrates and used for hydrogel preparation. After their encapsulation in hydrogels for 72 h, retrieved MSCs were analyzed for immunomodulatory activities, apoptosis, stem cell properties, senescence, CD9+, CD63+ and CD81+ extracellular vesicle (EV) release, and metabolism-related changes. PRP-hydrogels stimulated immunosuppressive functions of MSCs, along with their upregulated susceptibility to cell death in communication with PBMCs and augmented caspase 3/7 activity. We found impaired clonal growth and cell cycle progression, and more pronounced β-galactosidase activity as well as accumulation of LC3-II-positive vacuoles in PRP-MSCs. Stimuli derived from PRP-hydrogels upregulated AKT and reduced mTOR phosphorylation in MSCs, which suggests an initiation of survival-related processes. Our results showed that PRP-hydrogels might represent a metabolically stressful environment, inducing acidification of MSCs, reducing polarization of the mitochondrial membrane and increasing lipid accumulation. These features were not detected in FBR-MSCs, which showed reduced CD63+ and CD81+ EV production and maintained clonogenicity. Our data revealed that PRP-derived hematoma components cause metabolic adaptation of MSCs followed by increased immune regulatory functions. For the first time, we showed that PRP stimuli represent a survival challenge and “apoptotic priming” that are detrimental for stem cell-like growth of MSCs and important for their therapeutic consideration.
Graphical Abstract
Collapse
|
43
|
Florêncio KGD, Edson EA, Fernandes KSDS, Luiz JPM, Pinto FDCL, Pessoa ODL, Cunha FDQ, Machado-Neto JA, Wilke DV. Chromomycin A 5 induces bona fide immunogenic cell death in melanoma. Front Immunol 2022; 13:941757. [PMID: 36439184 PMCID: PMC9682167 DOI: 10.3389/fimmu.2022.941757] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 10/12/2022] [Indexed: 08/27/2023] Open
Abstract
PURPOSE Some first-line cytotoxic chemotherapics, e.g. doxorubicin, paclitaxel and oxaliplatin, induce activation of the immune system through immunogenic cell death (ICD). Tumor cells undergoing ICD function as a vaccine, releasing damage-associated molecular patterns (DAMPs), which act as adjuvants, and neoantigens of the tumor are recognized as antigens. ICD induction is rare, however it yields better and long-lasting antitumor responses to chemotherapy. Advanced metastatic melanoma (AMM) is incurable for more than half of patients. The discovery of ICD inducers against AMM is an interesting drug discovery strategy with high translational potential. Here we evaluated ICD induction of four highly cytotoxic chromomycins A (CA5-8). METHODS ICD features and DAMPs were evaluated using several in vitro techniques with metastatic melanoma cell line (B16-F10) exposed to chromomcins A5-8 such as flow cytometry, western blot, RT-PCR and luminescence. Additionally in vivo vaccination assays with CA5-treated cells in a syngeneic murine model (C57Bl/6) were performed to confirm ICD evaluating the immune cells activation and their antitumor activity. RESULTS B16-F10 treated with CA5-8 and doxorubicin exhibited ICD features such as autophagy and apoptosis, externalization of calreticulin, and releasing of HMGB1. However, CA5-treated cells had the best profile, also inducing ATP release, ERp57 externalization, phosphorylation of eIF2α and altering expression of transcription of genes related to autophagy, endoplasmic reticulum stress, and apoptosis. Bona fide ICD induction by CA5 was confirmed by vaccination of C57BL/6 mice with CA5-treated cells which activated antigen-presenting cells and T lymphocytes and stimulated antitumor activity. CONCLUSION CA5 induces bona fide immunogenic cell death on melanoma.
Collapse
Affiliation(s)
- Katharine Gurgel Dias Florêncio
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil
| | - Evelline Araújo Edson
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil
| | - Keilla Santana da Silva Fernandes
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil
| | - João Paulo Mesquita Luiz
- Center for Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | | | - Fernando de Queiroz Cunha
- Center for Research in Inflammatory Diseases (CRID), Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | | | - Diego Veras Wilke
- Drug Research and Development Center, Department of Physiology and Pharmacology, School of Medicine, Federal University of Ceara, Ceara, Brazil
| |
Collapse
|
44
|
Ilha M, Meira Martins LA, da Silveira Moraes K, Dias CK, Thomé MP, Petry F, Rohden F, Borojevic R, Trindade VMT, Klamt F, Barbé‐Tuana F, Lenz G, Guma FCR. Caveolin-1 influences mitochondrial plasticity and function in hepatic stellate cell activation. Cell Biol Int 2022; 46:1787-1800. [PMID: 35971753 PMCID: PMC9804617 DOI: 10.1002/cbin.11876] [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: 01/15/2022] [Revised: 03/21/2022] [Accepted: 05/26/2022] [Indexed: 01/05/2023]
Abstract
Caveolin-1 (Cav-1) is an integral membrane protein present in all organelles, responsible for regulating and integrating multiple signals as a platform. Mitochondria are extremely adaptable to external cues in chronic liver diseases, and expression of Cav-1 may affect mitochondrial flexibility in hepatic stellate cells (HSCs) activation. We previously demonstrated that exogenous expression of Cav-1 was sufficient to increase some classical markers of activation in HSCs. Here, we aimed to evaluate the influence of exogenous expression and knockdown of Cav-1 on regulating the mitochondrial plasticity, metabolism, endoplasmic reticulum (ER)-mitochondria distance, and lysosomal activity in HSCs. To characterize the mitochondrial, lysosomal morphology, and ER-mitochondria distance, we perform transmission electron microscope analysis. We accessed mitochondria and lysosomal networks and functions through a confocal microscope and flow cytometry. The expression of mitochondrial machinery fusion/fission genes was examined by real-time polymerase chain reaction. Total and mitochondrial cholesterol content was measured using Amplex Red. To define energy metabolism, we used the Oroboros system in the cells. We report that GRX cells with exogenous expression or knockdown of Cav-1 changed mitochondrial morphometric parameters, OXPHOS metabolism, ER-mitochondria distance, lysosomal activity, and may change the activation state of HSC. This study highlights that Cav-1 may modulate mitochondrial function and structural reorganization in HSC activation, being a potential candidate marker for chronic liver diseases and a molecular target for therapeutic intervention.
Collapse
Affiliation(s)
- Mariana Ilha
- Programa de Pós‐Graduação em Ciências Biológicas‐Bioquímica, Instituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do Sul – UFRGSPorto AlegreRio Grande do SulBrasil,Department of Clinical Nutrition, Institute of Public Health and Clinical NutritionUniversity of Eastern FinlandKuopioFinland
| | - Leo A. Meira Martins
- Programa de Pós‐Graduação em Ciências Biológicas‐Bioquímica, Instituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do Sul – UFRGSPorto AlegreRio Grande do SulBrasil,Departamento de Fisiologia, Instituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do Sul ‐ UFRGSPorto AlegreRio Grande do SulBrasil
| | - Ketlen da Silveira Moraes
- Departamento de Bioquímica, Instituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do Sul ‐ UFRGSPorto AlegreRio Grande do SulBrasil
| | - Camila K. Dias
- Programa de Pós‐Graduação em Ciências Biológicas‐Bioquímica, Instituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do Sul – UFRGSPorto AlegreRio Grande do SulBrasil
| | - Marcos P. Thomé
- Departamento de Biofísica e Centro de BiotecnologiaUniversidade Federal do Rio Grande do Sul ‐ UFRGSPorto AlegreRio Grande do SulBrasil
| | - Fernanda Petry
- Programa de Pós‐Graduação em Ciências Biológicas‐Bioquímica, Instituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do Sul – UFRGSPorto AlegreRio Grande do SulBrasil
| | - Francieli Rohden
- Programa de Pós‐Graduação em Ciências Biológicas‐Bioquímica, Instituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do Sul – UFRGSPorto AlegreRio Grande do SulBrasil
| | - Radovan Borojevic
- Centro de Medicina RegenerativaFaculdade Arthur Sa Earp Neto ‐ Faculdade de Medicina de PetrópolisRio de JaneiroBrasil
| | - Vera M. T. Trindade
- Departamento de Bioquímica, Instituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do Sul ‐ UFRGSPorto AlegreRio Grande do SulBrasil
| | - Fábio Klamt
- Departamento de Bioquímica, Instituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do Sul ‐ UFRGSPorto AlegreRio Grande do SulBrasil
| | - Florência Barbé‐Tuana
- Programa de Pós‐Graduação em Biologia Celular e MolecularEscola de Ciências da Pontifícia Universidade Católica do Rio Grande do Sul‐ PUCRSPorto AlegreRio Grande do SulBrasil
| | - Guido Lenz
- Departamento de Biofísica e Centro de BiotecnologiaUniversidade Federal do Rio Grande do Sul ‐ UFRGSPorto AlegreRio Grande do SulBrasil
| | - Fátima C. R. Guma
- Departamento de Bioquímica, Instituto de Ciências Básicas da SaúdeUniversidade Federal do Rio Grande do Sul ‐ UFRGSPorto AlegreRio Grande do SulBrasil,Centro de Microscopia e MicroanáliseUniversidade Federal do Rio Grande do Sul ‐ UFRGSPorto AlegreRio Grande do SulBrasil
| |
Collapse
|
45
|
Ex Vivo Fluorescence Confocal Microscopy (FCM) Ensures Representative Tissue in Prostate Cancer Biobanking: A Feasibility Study. Int J Mol Sci 2022; 23:ijms232012103. [PMID: 36292970 PMCID: PMC9603154 DOI: 10.3390/ijms232012103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/24/2022] [Accepted: 09/30/2022] [Indexed: 12/05/2022] Open
Abstract
Background: Biobanking of prostate carcinoma is particularly challenging due to the actual cancer within the organ often without clear margins. Frozen sections are to date the only way to examine the biobank material for its tumor content. We used ex vivo fluorescence confocal microscopy (FCM) to analyze biobank samples prior to cryoasservation. Methods: 127 punch biopsies were acquired from prostatectomy-specimens from 40 patients. These biopsies were analyzed with a Vivascope 2500-G4 prior to their transfer to the biobank. In difficult cases, larger samples of the prostatectomy specimens were FCM scanned in order to locate tumor foci. After patient acquisition, all samples were taken from the biobank and analyzed. We compared the results of the FCM examinations with the results of conventional histology and measured the DNA content. Results: With upstream FCM, the tumor content of biobank samples could be determined with high confidence. The detection rate of representative biobank samples was increased due to the rapid feedback. The biobank samples were suitable for further molecular analysis. Conclusion: FCM allows for the first time lossless microscopic analysis of biobank samples prior to their cryoasservation and guarantees representative tumor and normal tissue for further molecular analysis.
Collapse
|
46
|
Hao W, Dian M, Zhou Y, Zhong Q, Pang W, Li Z, Zhao Y, Ma J, Lin X, Luo R, Li Y, Jia J, Shen H, Huang S, Dai G, Wang J, Sun Y, Xiao D. Autophagy induction promoted by m 6A reader YTHDF3 through translation upregulation of FOXO3 mRNA. Nat Commun 2022; 13:5845. [PMID: 36195598 PMCID: PMC9532426 DOI: 10.1038/s41467-022-32963-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 08/24/2022] [Indexed: 12/08/2022] Open
Abstract
Autophagy is crucial for maintaining cellular energy homeostasis and for cells to adapt to nutrient deficiency, and nutrient sensors regulating autophagy have been reported previously. However, the role of eiptranscriptomic modifications such as m6A in the regulation of starvation-induced autophagy is unclear. Here, we show that the m6A reader YTHDF3 is essential for autophagy induction. m6A modification is up-regulated to promote autophagosome formation and lysosomal degradation upon nutrient deficiency. METTL3 depletion leads to a loss of functional m6A modification and inhibits YTHDF3-mediated autophagy flux. YTHDF3 promotes autophagy by recognizing m6A modification sites around the stop codon of FOXO3 mRNA. YTHDF3 also recruits eIF3a and eIF4B to facilitate FOXO3 translation, subsequently initiating autophagy. Overall, our study demonstrates that the epitranscriptome regulator YTHDF3 functions as a nutrient responder, providing a glimpse into the post-transcriptional RNA modifications that regulate metabolic homeostasis.
Collapse
Affiliation(s)
- WeiChao Hao
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, 510080, Guangzhou, China
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - MeiJuan Dian
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, 510515, Guangzhou, China
| | - Ying Zhou
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, 510515, Guangzhou, China
| | - QiuLing Zhong
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - WenQian Pang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - ZiJian Li
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - YaYan Zhao
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, 510080, Guangzhou, China
| | - JiaCheng Ma
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, 10084, Beijing, China
| | - XiaoLin Lin
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 510315, Guangzhou, China
| | - RenRu Luo
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, 518107, Guangdong, China
| | - YongLong Li
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, 510515, Guangzhou, China
| | - JunShuang Jia
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - HongFen Shen
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
| | - ShiHao Huang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, 510515, Guangzhou, China
| | - GuanQi Dai
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, 510515, Guangzhou, China
| | - JiaHong Wang
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China.
| | - Yan Sun
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 510080, Guangzhou, China.
| | - Dong Xiao
- Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China.
- Institute of Comparative Medicine & Laboratory Animal Center, Southern Medical University, 510515, Guangzhou, China.
- National Demonstration Center for Experimental Education of Basic Medical Sciences, Southern Medical University, 510515, Guangzhou, China.
| |
Collapse
|
47
|
Qu X, Liu Z, Li N, Ma B, Zhao H, Li Y, Lei B, Du Y. Biodegradable biocompatible MgO/Eu nanodrug with Acid-Base conversion capacity for targeted lung cancer therapy. CHEMICAL ENGINEERING JOURNAL 2022; 446:136323. [DOI: 10.1016/j.cej.2022.136323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
|
48
|
Liu S, Jiang L, Miao H, Lv Y, Zhang Q, Ma M, Duan W, Huang Y, Wei X. Autophagy regulation of ATG13 and ATG27 on biofilm formation and antifungal resistance in Candida albicans. BIOFOULING 2022; 38:926-939. [PMID: 36476055 DOI: 10.1080/08927014.2022.2153332] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 10/20/2022] [Accepted: 11/25/2022] [Indexed: 06/17/2023]
Abstract
Autophagy is a highly conserved catabolic pathway that is vital for cells; however, the effects of autophagy on the biofilm formation and antifungal resistance of Candida albicans are still unknown. In this study, the potential molecular mechanisms of autophagy in biofilm formation and antifungal resistance were investigated. It was found that 3536 genes were differentially expressed between biofilm and planktonic C. albicans. ATG gene expression and autophagy activity were higher in biofilm than in planktonic C. albicans. Autophagic activities were higher in matured biofilms than that in pre-matured biofilms. Autophagy was involved in C. albicans biofilm formation and its activity increased during biofilm maturation. Further, ALP activity, AO staining cells, and autophagosomes inside cells were obviously reduced in biofilms of atg13Δ/Δ and atg27Δ/Δ strains; moreover, biofilm formation and antifungal resistance were also significantly decreased. Lastly, autophagy regulates biofilm formation and drug resistance of C. albicans and could be served as a new molecular target to the C. albicans biofilm infections.
Collapse
Affiliation(s)
- Siqi Liu
- Department of Operative Dentistry and Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Liuliu Jiang
- Department of Operative Dentistry and Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Haochen Miao
- Department of Operative Dentistry and Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Ying Lv
- Department of Operative Dentistry and Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Qinqin Zhang
- Department of Operative Dentistry and Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Ming Ma
- Department of Operative Dentistry and Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Wei Duan
- Department of Operative Dentistry and Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Yun Huang
- Department of Operative Dentistry and Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| | - Xin Wei
- Department of Operative Dentistry and Endodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing, China
| |
Collapse
|
49
|
Lacret R, Puerta A, Granica S, González-Bakker A, Hevia D, Teng Y, Sánchez-Mateo CC, Pérez de Paz PL, Padrón JM. Bioactive Potential: A Pharmacognostic Definition through the Screening of Four Hypericum Species from the Canary Islands. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27186101. [PMID: 36144833 PMCID: PMC9505652 DOI: 10.3390/molecules27186101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 11/30/2022]
Abstract
In this work, we propose a general methodology to assess the bioactive potential (BP) of extracts in the quest of vegetable-based drugs. To exemplify the method, we studied the anticancer potential (AP) of four endemic species of genus Hypericum (Hypericum canariense L, Hypericum glandulosum Aiton, Hypericum grandifolium Choisy and Hypericum reflexum L.f) from the Canary Islands. Microextracts were obtained from the aerial parts of these species and were tested against six human tumor cell lines, A549 (non-small-cell lung), HBL-100 (breast), HeLa (cervix), SW1573 (non-small-cell lung), T-47D (breast) and WiDr (colon). The methanol–water microextracts were evaluated further for cell migration, autophagy and cell death. The most promising bioactive polar microextracts were analyzed by UHPLC–DAD–MS. The extraction yield, the bioactivity evaluation and the chemical profiling by LC–MS suggested that H. grandifolium was the species with the highest AP. Label-free live-cell imaging studies on HeLa cells exposed to the methanol–water microextract of H. grandifolium enabled observing cell death and several apoptotic hallmarks. Overall, this study allows us to select Hypericum grandifolium Choisy as a source of new chemical entities with a potential interest for cancer treatment.
Collapse
Affiliation(s)
- Rodney Lacret
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO-AG), Universidad de La Laguna, Avda. Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain
- Departamento de Medicina Física y Farmacología, Facultad de Farmacia, Universidad de La Laguna, Tenerife, 38200 La Laguna, Spain
- Correspondence:
| | - Adrián Puerta
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO-AG), Universidad de La Laguna, Avda. Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain
| | - Sebastian Granica
- Microbiota Lab, Centre of Preclinical Studies, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland
| | - Aday González-Bakker
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO-AG), Universidad de La Laguna, Avda. Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain
| | - Danela Hevia
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO-AG), Universidad de La Laguna, Avda. Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain
| | - Yiling Teng
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO-AG), Universidad de La Laguna, Avda. Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain
| | - Candelaria C. Sánchez-Mateo
- Departamento de Medicina Física y Farmacología, Facultad de Farmacia, Universidad de La Laguna, Tenerife, 38200 La Laguna, Spain
| | - Pedro Luis Pérez de Paz
- Departamento de Botánica, Ecología y Fisiología Vegetal, Facultad de Farmacia, Universidad de La Laguna, Tenerife, 38200 La Laguna, Spain
| | - José M. Padrón
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO-AG), Universidad de La Laguna, Avda. Astrofísico Francisco Sánchez 2, 38206 La Laguna, Spain
| |
Collapse
|
50
|
Popovici V, Matei E, Cozaru GC, Bucur L, Gîrd CE, Schröder V, Ozon EA, Karampelas O, Musuc AM, Atkinson I, Rusu A, Petrescu S, Mitran RA, Anastasescu M, Caraiane A, Lupuliasa D, Aschie M, Badea V. Evaluation of Usnea barbata (L.) Weber ex F.H. Wigg Extract in Canola Oil Loaded in Bioadhesive Oral Films for Potential Applications in Oral Cavity Infections and Malignancy. Antioxidants (Basel) 2022; 11:antiox11081601. [PMID: 36009320 PMCID: PMC9404812 DOI: 10.3390/antiox11081601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 12/16/2022] Open
Abstract
Usnea lichens are known for their beneficial pharmacological effects with potential applications in oral medicine. This study aims to investigate the extract of Usnea barbata (L.) Weber ex F.H. Wigg from the Călimani Mountains in canola oil as an oral pharmaceutical formulation. In the present work, bioadhesive oral films (F-UBO) with U. barbata extract in canola oil (UBO) were formulated, characterized, and evaluated, evidencing their pharmacological potential. The UBO-loaded films were analyzed using standard methods regarding physicochemical and pharmacotechnical characteristics to verify their suitability for topical administration on the oral mucosa. F-UBO suitability confirmation allowed for the investigation of antimicrobial and anticancer potential. The antimicrobial properties against Staphylococcus aureus ATCC 25923, Pseudomonas aeruginosa ATCC 27353, Candida albicans ATCC 10231, and Candida parapsilosis ATCC 22019 were evaluated by a resazurin-based 96-well plate microdilution method. The brine shrimp lethality assay (BSL assay) was the animal model cytotoxicity prescreen, followed by flow cytometry analyses on normal blood cells and oral epithelial squamous cell carcinoma CLS-354 cell line, determining cellular apoptosis, caspase-3/7 activity, nuclear condensation and lysosomal activity, oxidative stress, cell cycle, and cell proliferation. The results indicate that a UBO-loaded bioadhesive film’s weight is 63 ± 1.79 mg. It contains 315 µg UBO, has a pH = 6.97 ± 0.01, a disintegration time of 124 ± 3.67 s, and a bioadhesion time of 86 ± 4.12 min, being suitable for topical administration on the oral mucosa. F-UBO showed moderate dose-dependent inhibitory effects on the growth of both bacterial and fungal strains. Moreover, in CLS-354 tumor cells, F-UBO increased oxidative stress, diminished DNA synthesis, and induced cell cycle arrest in G0/G1. All these properties led to considering UBO-loaded bioadhesive oral films as a suitable phytotherapeutic formulation with potential application in oral infections and neoplasia.
Collapse
Affiliation(s)
- Violeta Popovici
- Department of Microbiology and Immunology, Faculty of Dental Medicine, Ovidius University of Constanta, 7 Ilarie Voronca Street, 900684 Constanta, Romania
| | - Elena Matei
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, Ovidius University of Constanta, CEDMOG, 145 Tomis Blvd., 900591 Constanta, Romania
- Correspondence: (E.M.); (V.S.); (E.A.O.); (O.K.); (A.M.M.)
| | - Georgeta Camelia Cozaru
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, Ovidius University of Constanta, CEDMOG, 145 Tomis Blvd., 900591 Constanta, Romania
- Clinical Service of Pathology, Sf. Apostol Andrei Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania
| | - Laura Bucur
- Department of Pharmacognosy, Faculty of Pharmacy, Ovidius University of Constanta, 6 Capitan Al. Serbanescu Street, 900001 Constanta, Romania
| | - Cerasela Elena Gîrd
- Department of Pharmacognosy, Phytochemistry, and Phytotherapy, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| | - Verginica Schröder
- Department of Cellular and Molecular Biology, Faculty of Pharmacy, Ovidius University of Constanta, 6 Capitan Al. Serbanescu Street, 900001 Constanta, Romania
- Correspondence: (E.M.); (V.S.); (E.A.O.); (O.K.); (A.M.M.)
| | - Emma Adriana Ozon
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
- Correspondence: (E.M.); (V.S.); (E.A.O.); (O.K.); (A.M.M.)
| | - Oana Karampelas
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
- Correspondence: (E.M.); (V.S.); (E.A.O.); (O.K.); (A.M.M.)
| | - Adina Magdalena Musuc
- “Ilie Murgulescu” Institute of Physical Chemistry, Romanian Academy, 202 Spl. Independentei, 060021 Bucharest, Romania
- Correspondence: (E.M.); (V.S.); (E.A.O.); (O.K.); (A.M.M.)
| | - Irina Atkinson
- “Ilie Murgulescu” Institute of Physical Chemistry, Romanian Academy, 202 Spl. Independentei, 060021 Bucharest, Romania
| | - Adriana Rusu
- “Ilie Murgulescu” Institute of Physical Chemistry, Romanian Academy, 202 Spl. Independentei, 060021 Bucharest, Romania
| | - Simona Petrescu
- “Ilie Murgulescu” Institute of Physical Chemistry, Romanian Academy, 202 Spl. Independentei, 060021 Bucharest, Romania
| | - Raul-Augustin Mitran
- “Ilie Murgulescu” Institute of Physical Chemistry, Romanian Academy, 202 Spl. Independentei, 060021 Bucharest, Romania
| | - Mihai Anastasescu
- “Ilie Murgulescu” Institute of Physical Chemistry, Romanian Academy, 202 Spl. Independentei, 060021 Bucharest, Romania
| | - Aureliana Caraiane
- Department of Oral Rehabilitation, Faculty of Dental Medicine, Ovidius University of Constanta, 7 Ilarie Voronca Street, 900684 Constanta, Romania
| | - Dumitru Lupuliasa
- Department of Pharmaceutical Technology and Biopharmacy, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| | - Mariana Aschie
- Center for Research and Development of the Morphological and Genetic Studies of Malignant Pathology, Ovidius University of Constanta, CEDMOG, 145 Tomis Blvd., 900591 Constanta, Romania
- Clinical Service of Pathology, Sf. Apostol Andrei Emergency County Hospital, 145 Tomis Blvd., 900591 Constanta, Romania
| | - Victoria Badea
- Department of Microbiology and Immunology, Faculty of Dental Medicine, Ovidius University of Constanta, 7 Ilarie Voronca Street, 900684 Constanta, Romania
| |
Collapse
|