1
|
Cao B, Gu S, Shen Z, Zhang Y, Shen Y. Construction of lysosome-related prognostic signature to predict the survival outcomes and selecting suitable drugs for patients with HNSCC. Biofactors 2024. [PMID: 39495139 DOI: 10.1002/biof.2140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
Lysosomes are digestive organelles responsible for endocytosis and autophagy. Recently, the malignancy and invasiveness head and neck squamous cell carcinoma (HNSCC) has been increasingly studied with the role of lysosomes. A list of lysosome-related genes were obtained from MSigDB. A Spearman correlation and univariate Cox regression analyses combined with differential expression analysis were conducted to detect differentially expressed lysosome-related genes related to prognosis. The prediction of prognostic signature was evaluated by plotting survival curve, ROC, and by developing a nomogram. Immune subtypes, infiltration of immune cells, GSVA, TIDE, IC50 of common chemotherapy and targeted therapy, GO, and KEGG function enrichment analyses were carried out to explore the immune microenvironment of the signature. We constructed a lysosome-related prognostic signature that could function as an independent prognostic indicator for patients with HNSCC. High-risk patients were better suited to receive Doxorubicin, Mitomycin C, Pyrimethamine, anti-PD-L1 and anti-CTLA-4 immunotherapy, whereas low-risk patients had sensitivity to Lapatinib. GO functional enrichment analysis showed that prognostic features were strongly associated with epidermis-related functions (e.g., epidermal cell differentiation, epidermal development, and keratinization). In addition, a KEGG function enrichment analysis revealed a potential relationship between the risk assessment model and cardiomyopathy. We constructed a prognostic signature based on lysosome-related genes and successfully predicted the survival outcome of HNSCC patients, which not only provides potential guidance for personalized treatment but also provides a new idea for precision treatment of HNSCC.
Collapse
Affiliation(s)
- Bing Cao
- Department of Otorhinolaryngology Head and Neck Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Shanshan Gu
- Department of Otorhinolaryngology Head and Neck Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Zhisen Shen
- Department of Otorhinolaryngology Head and Neck Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Yuna Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Lihuili Hospital Affiliated to Ningbo University, Ningbo, China
| | - Yiming Shen
- Department of Otology and Skull Base Surgery, National Health Commission Key Laboratory of Hearing Medicine (Fudan University), Shanghai, China
| |
Collapse
|
2
|
Shi Z, Zeng Y, Luo J, Wang X, Ma G, Zhang T, Huang P. Endogenous Magnetic Lipid Droplet-Mediated Cascade-Targeted Sonodynamic Therapy as an Approach to Reversing Breast Cancer Multidrug Resistance. ACS NANO 2024; 18:28659-28674. [PMID: 39387174 DOI: 10.1021/acsnano.4c05938] [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: 10/12/2024]
Abstract
Multidrug resistance (MDR) has emerged as a major barrier to effective breast cancer treatment, contributing to high rates of chemotherapy failure and disease recurrence. There is thus a pressing need to overcome MDR and to facilitate the efficient and precise treatment of breast cancer in a targeted manner. In this study, endogenous functional lipid droplets (IR780@LDs-Fe3O4/OA) were developed and used to effectively overcome the limited diffusion distance of reactive oxygen species owing to their amenability to cascade-targeted delivery, thereby facilitating precise and effective sonodynamic therapy (SDT) for MDR breast cancer. Initially, IR780@LDs-Fe3O4/OA was efficiently enriched within tumor sites in a static magnetic field, achieving the visualization of tumor treatment. Subsequently, the cascade-targeted SDT combined with the Fenton effect induced lysosome membrane permeabilization and relieved lysosomal sequestration, thus elevating drug concentration at the target site. This treatment approach also suppressed ATP production, thereby inhibiting P-glycoprotein-mediated chemotherapeutic drug efflux. This cascade-targeted SDT strategy significantly increased the sensitivity of MDR cells to doxorubicin, increasing the IC50 value of doxorubicin by approximately 10-fold. Moreover, the cascade-targeted SDT also altered the gene expression profiles of MDR cells and suppressed the expression of MDR-related genes. In light of these promising results, the combination of cascade-targeted SDT and conventional chemotherapy holds great clinical promise as an effective treatment modality with excellent biocompatibility that can improve MDR breast cancer patient outcomes.
Collapse
Affiliation(s)
- Zhan Shi
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
| | - Yiqing Zeng
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
| | - Jiali Luo
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
| | - Xue Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
| | - Guangrong Ma
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
| | - Tao Zhang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, No. 88 Jiefang Road, Shangcheng District, Hangzhou 310009, P. R. China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, No. 66 Dongxin Avenue, Binjiang District, Hangzhou 310053, P. R. China
| |
Collapse
|
3
|
Yang L, Meng B, Gong X, Jiang Y, Shentu X, Xue Z. Investigation of the synergistic effect mechanism underlying sequential use of palbociclib and cisplatin through integral proteomic and glycoproteomic analysis. Anticancer Drugs 2024; 35:806-816. [PMID: 39011652 PMCID: PMC11392100 DOI: 10.1097/cad.0000000000001633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Chemoresistance largely hampers the clinical use of chemodrugs for cancer patients, combination or sequential drug treatment regimens have been designed to minimize chemotoxicity and resensitize chemoresistance. In this work, the cytotoxic effect of cisplatin was found to be enhanced by palbociclib pretreatment in HeLa cells. With the integration of liquid chromatography-mass spectrometry-based proteomic and N-glycoproteomic workflow, we found that palbociclib alone mainly enhanced the N-glycosylation alterations in HeLa cells, while cisplatin majorly increased the different expression proteins related to apoptosis pathways. As a result, the sequential use of two drugs induced a higher expression level of apoptosis proteins BAX and BAK. Those altered N-glycoproteins induced by palbociclib were implicated in pathways that were closely associated with cell membrane modification and drug sensitivity. Specifically, the top four frequently glycosylated proteins FOLR1, L1CAM, CD63, and LAMP1 were all associated with drug resistance or drug sensitivity. It is suspected that palbociclib-induced N-glycosylation on the membrane protein allowed the HeLa cell to become more vulnerable to cisplatin treatment. Our study provides new insights into the mechanisms underlying the sequential use of target drugs and chemotherapy drugs, meanwhile suggesting a high-efficiency approach that involves proteomic and N-glycoproteomic to facilitate drug discovery.
Collapse
Affiliation(s)
- Lulu Yang
- Faculty of Life Sciences, China Jiliang University, Hangzhou
| | - Bo Meng
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - Xiaoyun Gong
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - You Jiang
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| | - Xuping Shentu
- Faculty of Life Sciences, China Jiliang University, Hangzhou
| | - Zhichao Xue
- Technology Innovation Center of Mass Spectrometry for State Market Regulation, Center for Advanced Measurement Science, National Institute of Metrology, Beijing, China
| |
Collapse
|
4
|
Tian Y, Wang X, Wu C, Qiao J, Jin H, Li H. A protracted war against cancer drug resistance. Cancer Cell Int 2024; 24:326. [PMID: 39342202 PMCID: PMC11439304 DOI: 10.1186/s12935-024-03510-2] [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: 04/11/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
Currently, even the most effective anti-cancer therapies are often limited by the development of drug resistance and tumor relapse, which is a major challenge facing current cancer research. A deep understanding of the molecular and biochemical bases of drug efficacy that can help predict the clinical drug resistance, coupled with the evolution of systematic genomic and proteomic technologies, have facilitated studies identifying and elucidating the underlying mechanisms. In this review, we focus on several important issues on cancer drug resistance and provide a framework for understanding the common ways by which cancers develop resistance to therapeutic agents. With the increasing arsenal of novel anticancer agents and techniques, there are now unprecedented opportunities to understand and overcome drug resistance. The proteolysis targeting chimera (PROTAC) technology, immunotherapy, nanomedicine, and real-time monitoring of drug response all provide effective approaches for combating drug resistance. In addition to the advancement of therapeutic technologies, the revolution of treatment concept is also of great importance. We can take advantage of the interplay between drug sensitive and resistant subclones for combating cancer. However, there remains a long way to go in the protracted war against cancer drug resistance.
Collapse
Affiliation(s)
- Yuan Tian
- School of Lifesciences, Shanghai University, 333 Nanchen Road, Shanghai, 200444, P.R. China
| | - Xiaowei Wang
- Department of Thoracic Surgery/Clinical Research Center, The First Affiliated Hospital of Navy Medical University, 168 Changhai Road, Shanghai, 200433, P.R. China
| | - Cong Wu
- Department of Thoracic Surgery/Clinical Research Center, The First Affiliated Hospital of Navy Medical University, 168 Changhai Road, Shanghai, 200433, P.R. China
| | - Jiaming Qiao
- School of Lifesciences, Shanghai University, 333 Nanchen Road, Shanghai, 200444, P.R. China
| | - Hai Jin
- Department of Thoracic Surgery/Clinical Research Center, The First Affiliated Hospital of Navy Medical University, 168 Changhai Road, Shanghai, 200433, P.R. China.
| | - Huafei Li
- School of Lifesciences, Shanghai University, 333 Nanchen Road, Shanghai, 200444, P.R. China.
| |
Collapse
|
5
|
Dinić J, Podolski-Renić A, Novaković M, Li L, Opsenica I, Pešić M. Plant-Based Products Originating from Serbia That Affect P-glycoprotein Activity. Molecules 2024; 29:4308. [PMID: 39339303 PMCID: PMC11433820 DOI: 10.3390/molecules29184308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/02/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Our review paper evaluates the impact of plant-based products, primarily derived from plants from Serbia, on P-glycoprotein (P-gp) activity and their potential in modulating drug resistance in cancer therapy. We focus on the role and regulation of P-gp in cellular physiology and its significance in addressing multidrug resistance in cancer therapy. Additionally, we discuss the modulation of P-gp activity by 55 natural product drugs, including derivatives for some of them, based on our team's research findings since 2011. Specifically, we prospect into sesquiterpenoids from the genera Artemisia, Curcuma, Ferula, Inula, Petasites, and Celastrus; diterpenoids from the genera Salvia and Euphorbia; chalcones from the genera Piper, Glycyrrhiza, Cullen, Artemisia, and Humulus; riccardins from the genera Lunularia, Monoclea, Dumortiera, Plagiochila, and Primula; and diarylheptanoids from the genera Alnus and Curcuma. Through comprehensive analysis, we aim to highlight the potential of natural products mainly identified in plants from Serbia in influencing P-gp activity and overcoming drug resistance in cancer therapy, while also providing insights into future perspectives in this field.
Collapse
Affiliation(s)
- Jelena Dinić
- Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Despota Stefana 142, 11108 Belgrade, Serbia; (J.D.); (A.P.-R.)
| | - Ana Podolski-Renić
- Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Despota Stefana 142, 11108 Belgrade, Serbia; (J.D.); (A.P.-R.)
| | - Miroslav Novaković
- Institute of Chemistry, Technology and Metallurgy—National Institute of the Republic of Serbia, University of Belgrade, Njegoševa 12, 11000 Belgrade, Serbia;
| | - Liang Li
- Key Laboratory of Bioactive Substance and Function of Natural Medicines, State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Tiantan Xili, Beijing 100050, China;
| | - Igor Opsenica
- Faculty of Chemistry, University of Belgrade, Studentski trg 12–16, 11158 Belgrade, Serbia;
| | - Milica Pešić
- Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Despota Stefana 142, 11108 Belgrade, Serbia; (J.D.); (A.P.-R.)
| |
Collapse
|
6
|
Ombredane AS, Martins NO, de Souza GMV, Araujo VHS, Szlachetka ÍO, da Silva SW, da Rocha MCO, de Oliveira AS, Holanda CA, Romeiro LAS, Damas EBDO, Azevedo RB, Joanitti GA. Combinatory Effect of Pequi Oil ( Caryocar brasiliense)-Based Nanoemulsions Associated to Docetaxel and Anacardic Acid ( Anacardium occidentale) in Triple-Negative Breast Cancer Cells In Vitro. Pharmaceutics 2024; 16:1170. [PMID: 39339206 PMCID: PMC11435098 DOI: 10.3390/pharmaceutics16091170] [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: 07/01/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/30/2024] Open
Abstract
Combination therapy integrated with nanotechnology offers a promising alternative for breast cancer treatment. The inclusion of pequi oil, anacardic acid (AA), and docetaxel (DTX) in a nanoemulsion can amplify the antitumor effects of each molecule while reducing adverse effects. Therefore, the study aims to develop pequi oil-based nanoemulsions (PeNE) containing DTX (PDTX) or AA (PAA) and to evaluate their cytotoxicity against triple-negative breast cancer cells (4T1) in vitro. The PeNE without and with AA (PAA) and DTX (PDTX) were prepared by sonication and characterized by ZetaSizer® and electronic transmission microscopy. Viability testing and combination index (CI) were determined by MTT and Chou-Talalay methods, respectively. Flow cytometry was employed to investigate the effects of the formulations on cell structures. PeNE, PDTX, and PAA showed hydrodynamic diameter < 200 nm and a polydispersity index (PdI) of 0.3. The association PDTX + PAA induced a greater decrease in cell viability (~70%, p < 0.0001) and additive effect (CI < 1). In parallel, an association of the DTX + AA molecules led to antagonism (CI > 1). Additionally, PDTX + PAA induced an expressive morphological change, a major change in lysosome membrane permeation and mitochondria membrane permeation, cell cycle blockage in G2/M, and phosphatidylserine exposure. The study highlights the successful use of pequi oil nanoemulsions as delivery systems for DTX and AA, which enhances their antitumor effects against breast cancer cells. This nanotechnological approach shows significant potential for the treatment of triple-negative breast cancer.
Collapse
Affiliation(s)
- Alicia Simalie Ombredane
- Laboratory of Bioactive Compounds and Nanobiotechnology (LCBNano), University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasilia 72220-275, Brazil; (A.S.O.); (N.O.M.); (G.M.V.d.S.); (V.H.S.A.); (E.B.d.O.D.)
- Post-Graduation Program in Nanoscience and Nanobiotechnology, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasilia 70910-900, Brazil; (Í.O.S.); (S.W.d.S.); (M.C.O.d.R.); (R.B.A.)
| | - Natália Ornelas Martins
- Laboratory of Bioactive Compounds and Nanobiotechnology (LCBNano), University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasilia 72220-275, Brazil; (A.S.O.); (N.O.M.); (G.M.V.d.S.); (V.H.S.A.); (E.B.d.O.D.)
| | - Gabriela Mara Vieira de Souza
- Laboratory of Bioactive Compounds and Nanobiotechnology (LCBNano), University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasilia 72220-275, Brazil; (A.S.O.); (N.O.M.); (G.M.V.d.S.); (V.H.S.A.); (E.B.d.O.D.)
| | - Victor Hugo Sousa Araujo
- Laboratory of Bioactive Compounds and Nanobiotechnology (LCBNano), University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasilia 72220-275, Brazil; (A.S.O.); (N.O.M.); (G.M.V.d.S.); (V.H.S.A.); (E.B.d.O.D.)
| | - Ísis O. Szlachetka
- Post-Graduation Program in Nanoscience and Nanobiotechnology, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasilia 70910-900, Brazil; (Í.O.S.); (S.W.d.S.); (M.C.O.d.R.); (R.B.A.)
- Laboratory of Optical Espectroscopy, Physics Institute, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasilia 70910-900, Brazil
| | - Sebastião William da Silva
- Post-Graduation Program in Nanoscience and Nanobiotechnology, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasilia 70910-900, Brazil; (Í.O.S.); (S.W.d.S.); (M.C.O.d.R.); (R.B.A.)
- Laboratory of Optical Espectroscopy, Physics Institute, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasilia 70910-900, Brazil
| | - Márcia Cristina Oliveira da Rocha
- Post-Graduation Program in Nanoscience and Nanobiotechnology, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasilia 70910-900, Brazil; (Í.O.S.); (S.W.d.S.); (M.C.O.d.R.); (R.B.A.)
| | - Andressa Souza de Oliveira
- Graduate Program in Pharmaceutical Sciences, Department of Pharmacy, Health Sciences Faculty, University of Brasilia, Brasilia 70910-900, Brazil; (A.S.d.O.); (C.A.H.); (L.A.S.R.)
- Laboratory of Development of Therapeutic Innovations (LDT), Center for Tropical Medicine, Faculty of Medicine, University of Brasilia, Brasilia 70910-900, Brazil
| | - Cleonice Andrade Holanda
- Graduate Program in Pharmaceutical Sciences, Department of Pharmacy, Health Sciences Faculty, University of Brasilia, Brasilia 70910-900, Brazil; (A.S.d.O.); (C.A.H.); (L.A.S.R.)
- Laboratory of Development of Therapeutic Innovations (LDT), Center for Tropical Medicine, Faculty of Medicine, University of Brasilia, Brasilia 70910-900, Brazil
| | - Luiz Antonio Soares Romeiro
- Graduate Program in Pharmaceutical Sciences, Department of Pharmacy, Health Sciences Faculty, University of Brasilia, Brasilia 70910-900, Brazil; (A.S.d.O.); (C.A.H.); (L.A.S.R.)
- Laboratory of Development of Therapeutic Innovations (LDT), Center for Tropical Medicine, Faculty of Medicine, University of Brasilia, Brasilia 70910-900, Brazil
| | - Elysa Beatriz de Oliveira Damas
- Laboratory of Bioactive Compounds and Nanobiotechnology (LCBNano), University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasilia 72220-275, Brazil; (A.S.O.); (N.O.M.); (G.M.V.d.S.); (V.H.S.A.); (E.B.d.O.D.)
| | - Ricardo Bentes Azevedo
- Post-Graduation Program in Nanoscience and Nanobiotechnology, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasilia 70910-900, Brazil; (Í.O.S.); (S.W.d.S.); (M.C.O.d.R.); (R.B.A.)
| | - Graziella Anselmo Joanitti
- Laboratory of Bioactive Compounds and Nanobiotechnology (LCBNano), University of Brasilia, Campus Universitário—Centro Metropolitano, Ceilandia Sul, Brasilia 72220-275, Brazil; (A.S.O.); (N.O.M.); (G.M.V.d.S.); (V.H.S.A.); (E.B.d.O.D.)
- Post-Graduation Program in Nanoscience and Nanobiotechnology, University of Brasilia, Campus Universitário Darcy Ribeiro, Brasilia 70910-900, Brazil; (Í.O.S.); (S.W.d.S.); (M.C.O.d.R.); (R.B.A.)
| |
Collapse
|
7
|
Liu X, Li M, Woo S. Subcellular Drug Distribution: Exploring Organelle-Specific Characteristics for Enhanced Therapeutic Efficacy. Pharmaceutics 2024; 16:1167. [PMID: 39339204 PMCID: PMC11434838 DOI: 10.3390/pharmaceutics16091167] [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: 08/12/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
The efficacy and potential toxicity of drug treatments depends on the drug concentration at its site of action, intricately linked to its distribution within diverse organelles of mammalian cells. These organelles, including the nucleus, endosome, lysosome, mitochondria, endoplasmic reticulum, Golgi apparatus, lipid droplets, exosomes, and membrane-less structures, create distinct sub-compartments within the cell, each with unique biological features. Certain structures within these sub-compartments possess the ability to selectively accumulate or exclude drugs based on their physicochemical attributes, directly impacting drug efficacy. Under pathological conditions, such as cancer, many cells undergo dynamic alterations in subcellular organelles, leading to changes in the active concentration of drugs. A mechanistic and quantitative understanding of how organelle characteristics and abundance alter drug partition coefficients is crucial. This review explores biological factors and physicochemical properties influencing subcellular drug distribution, alongside strategies for modulation to enhance efficacy. Additionally, we discuss physiologically based computational models for subcellular drug distribution, providing a quantifiable means to simulate and predict drug distribution at the subcellular level, with the potential to optimize drug development strategies.
Collapse
Affiliation(s)
- Xin Liu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY 14214-8033, USA;
| | - Miaomiao Li
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH 43210-1267, USA;
| | - Sukyung Woo
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY 14214-8033, USA;
| |
Collapse
|
8
|
Ji N, Li H, Zhang Y, Li Y, Wang P, Chen X, Liu YN, Wang JQ, Yang Y, Chen ZS, Li Y, Wang R, Kong D. Lansoprazole (LPZ) reverses multidrug resistance (MDR) in cancer through impeding ATP-binding cassette (ABC) transporter-mediated chemotherapeutic drug efflux and lysosomal sequestration. Drug Resist Updat 2024; 76:101100. [PMID: 38885537 DOI: 10.1016/j.drup.2024.101100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/21/2024] [Accepted: 05/30/2024] [Indexed: 06/20/2024]
Abstract
AIMS Lansoprazole is one of the many proton pump inhibitors (PPIs) that acts more strongly with ABCB1 and ABCG2. The present study is to investigate the potential of lansoprazole on reversal of ABCB1/G2-mediated MDR in cancer, in vitro and in vivo. METHODS Reversal studies and combination evaluation were conducted to determine the synergistic anti-MDR effects on lansoprazole. Lysosomal staining was used to determination of lansoprazole on ABCB1-mediated lysosomal sequestration. Substrate accumulation and efflux assays, ATPase activity, and molecular docking were conducted to evaluate lansoprazole on ABCB1/G2 functions. Western blot and immunofluorescence were used to detect lansoprazole on ABCB1/G2 expression and subcellular localization. MDR nude mice models were established to evaluate the effects of lansoprazole on MDR in vivo. RESULTS Lansoprazole attenuated ABCB1/G2-mediated MDR and exhibited synergistic effects with substrate drugs in MDR cells. In vivo experiments demonstrated that lansoprazole attenuated ABCB1/G2-mediated MDR and exhibited synergistic effects that augmented the sensitivity of substrate anticancer drugs in ABCB1/G2-mediated settings without obvious toxicity. Lansoprazole impeded lysosomal sequestration mediated by ABCB1, leading to a substantial increase in intracellular accumulation of substrate drugs. The effects of lansoprazole were not attributable to downregulation or alterations in subcellular localization of ABCB1/G2. Lansoprazole promoted the ATPase activity of ABCB1/G2 and competitively bound to the substrate-binding region of ABCB1/G2. CONCLUSIONS These findings present novel therapeutic avenues whereby the combination of lansoprazole and chemotherapeutic agents mitigates MDR mediated by ABCB1/G2 overexpression.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Antineoplastic Agents/pharmacology
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- Cell Line, Tumor
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Neoplasm/drug effects
- Lansoprazole/pharmacology
- Lysosomes/metabolism
- Lysosomes/drug effects
- Mice, Nude
- Molecular Docking Simulation
- Neoplasm Proteins
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Neoplasms/pathology
- Proton Pump Inhibitors/pharmacology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Ning Ji
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin 300060, China; Tianjin Medical University, Tianjin 300070, China
| | - Hui Li
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin 300060, China; Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Yixuan Zhang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin 300060, China; Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Yuelin Li
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin 300060, China; Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Peiyu Wang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin 300060, China; Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Xin Chen
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin 300060, China; Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Yi-Nan Liu
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin 300060, China; Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Yueguo Li
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin 300060, China; Tianjin Medical University, Tianjin 300070, China.
| | - Ran Wang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin 300060, China; Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China.
| | - Dexin Kong
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, National Key Laboratory of Druggability Evaluation and Systematic Translational Medicine, Tianjin 300060, China; Tianjin Medical University, Tianjin 300070, China; Key Laboratory of Immune Microenvironment and Diseases (Ministry of Education), Tianjin Medical University, Tianjin 300070, China.
| |
Collapse
|
9
|
Li Z, Tan W, Li X, Wang Y, Dang Z, Zhang Z, Guan S, Zhu S, Li F, Zhang M. Unlocking lysosomal acidity to activate membranolytic module for accurately cancer theranostics. Bioorg Chem 2024; 153:107764. [PMID: 39232344 DOI: 10.1016/j.bioorg.2024.107764] [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: 02/17/2024] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/06/2024]
Abstract
Chemotherapy drug efflux, toxic side effects, and low efficacy against drug-resistant cells have plagued safe and efficient cancer theranostics. However, the materials or methods that resolve these defects all-in-one are scarce. Here, a new cancer theranostics strategy is proposed by utilizing changes in lysosomal acidity in cancer cells to activate the membranolytic model to overcome these obstacles together. Therefore, a simple fluorescent anthracene derivative Lyso-Mito is developed, which has a perfect pKa (4.62) value that falls between the pH of lysosomes in cancer and normal cells. Lyso-Mito itself can precisely target and convert the pH perturbation of lysosomes in cancer cells to fluorescent response and membranolytic module activity to accomplish the low drug efflux, weak toxic side effects, and low drug-resistant cancer diagnosis and treatment without linking other functional units or any additional assistance. Hereby, a new cancer theranostics strategy of integrating organelle microenvironment and the membranolytic model is realized.
Collapse
Affiliation(s)
- Zhuo Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Wenjia Tan
- China-Japan Union Hospital of Jilin University, Changchun 130041, China
| | - Xinru Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - YaJun Wang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zetao Dang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Zhaoxia Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Shuwen Guan
- College of Life Science, Jilin University, Changchun 130012, China
| | - Shoujun Zhu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China
| | - Feng Li
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China.
| | - Ming Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, China.
| |
Collapse
|
10
|
Agrawal HG, Khatun S, Rengan AK, Mishra AK. Tuning the Flavin Core via Donor Appendage for Selective Subcellular Bioimaging and PDT Application. Chemistry 2024; 30:e202401483. [PMID: 38853431 DOI: 10.1002/chem.202401483] [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: 04/16/2024] [Revised: 05/31/2024] [Accepted: 06/05/2024] [Indexed: 06/11/2024]
Abstract
Herein, we report a novel flavin analogue as singular chemical component for lysosome bioimaging, and inherited photosensitizer capability of the flavin core was demonstrated as a promising candidate for photodynamic therapy (PDT) application. Fine-tuning the flavin core with the incorporation of methoxy naphthyl appendage provides an appropriate chemical design, thereby offering photostability, selectivity, and lysosomal colocalization, along with the aggregation-induced emissive nature, making it suitable for lysosomal bioimaging applications. Additionally, photosensitization capability of the flavin core with photostable nature of the synthesized analogue has shown remarkable capacity for generating reactive oxygen species (ROS) within cells, making it a promising candidate for photodynamic therapy (PDT) application.
Collapse
Affiliation(s)
- Harsha Gopal Agrawal
- Department of Chemistry, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, 502285, India
| | - Sajmina Khatun
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, 502285, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, 502285, India
| | - Ashutosh Kumar Mishra
- Department of Chemistry, Indian Institute of Technology, Hyderabad, Sangareddy, Telangana, 502285, India
| |
Collapse
|
11
|
Akman M, Monteleone C, Doronzo G, Godel M, Napoli F, Merlini A, Campani V, Nele V, Balmas E, Chontorotzea T, Fontana S, Digiovanni S, Barbu FA, Astanina E, Jafari N, Salaroglio IC, Kopecka J, De Rosa G, Mohr T, Bertero A, Righi L, Novello S, Scagliotti GV, Bussolino F, Riganti C. TFEB controls sensitivity to chemotherapy and immuno-killing in non-small cell lung cancer. J Exp Clin Cancer Res 2024; 43:219. [PMID: 39107857 PMCID: PMC11304671 DOI: 10.1186/s13046-024-03142-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND In non-small cell lung cancer (NSCLC) the efficacy of chemo-immunotherapy is affected by the high expression of drug efflux transporters as ABCC1 and by the low expression of ABCA1, mediating the isopentenyl pyrophosphate (IPP)-dependent anti-tumor activation of Vγ9Vδ2 T-lymphocytes. In endothelial cells ABCA1 is a predicted target of the transcription factor EB (TFEB), but no data exists on the correlation between TFEB and ABC transporters involved in the chemo-immuno-resistance in NSCLC. METHODS The impact of TFEB/ABCC1/ABCA1 expression on NSCLC patients' survival was analyzed in the TCGA-LUAD cohort and in a retrospective cohort of our institution. Human NSCLC cells silenced for TFEB (shTFEB) were analyzed for ABC transporter expression, chemosensitivity and immuno-killing. The chemo-immuno-sensitizing effects of nanoparticles encapsulating zoledronic acid (NZ) on shTFEB tumors and on tumor immune-microenvironment were evaluated in Hu-CD34+ mice by single-cell RNA-sequencing. RESULTS TFEBlowABCA1lowABCC1high and TFEBhighABCA1highABCC1low NSCLC patients had the worst and the best prognosis, respectively, in the TCGA-LUAD cohort and in a retrospective cohort of patients receiving platinum-based chemotherapy or immunotherapy as first-line treatment. By silencing shTFEB in NSCLC cells, we demonstrated that TFEB was a transcriptional inducer of ABCA1 and a repressor of ABCC1. shTFEB cells had also a decreased activity of ERK1/2/SREBP2 axis, implying reduced synthesis and efflux via ABCA1 of cholesterol and its intermediate IPP. Moreover, TFEB silencing reduced cholesterol incorporation in mitochondria: this event increased the efficiency of OXPHOS and the fueling of ABCC1 by mitochondrial ATP. Accordingly, shTFEB cells were less immuno-killed by the Vγ9Vδ2 T-lymphocytes activated by IPP and more resistant to cisplatin. NZ, which increased IPP efflux but not OXPHOS and ATP production, sensitized shTFEB immuno-xenografts, by reducing intratumor proliferation and increasing apoptosis in response to cisplatin, and by increasing the variety of anti-tumor infiltrating cells (Vγ9Vδ2 T-lymphocytes, CD8+T-lymphocytes, NK cells). CONCLUSIONS This work suggests that TFEB is a gatekeeper of the sensitivity to chemotherapy and immuno-killing in NSCLC, and that the TFEBlowABCA1lowABCC1high phenotype can be predictive of poor response to chemotherapy and immunotherapy. By reshaping both cancer metabolism and tumor immune-microenvironment, zoledronic acid can re-sensitize TFEBlow NSCLCs, highly resistant to chemo- and immunotherapy.
Collapse
Affiliation(s)
- Muhlis Akman
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Ciro Monteleone
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Gabriella Doronzo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- IRCCS Candiolo Cancer Institute, Candiolo, Italy
| | - Martina Godel
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Francesca Napoli
- Pathology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Torino, Italy
| | - Alessandra Merlini
- Thoracic Oncology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Torino, Italy
| | - Virginia Campani
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy
| | - Valeria Nele
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy
| | - Elisa Balmas
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Tatiana Chontorotzea
- Center for Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Simona Fontana
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Sabrina Digiovanni
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Francesca Alice Barbu
- Pathology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Torino, Italy
| | | | - Niloufar Jafari
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Iris Chiara Salaroglio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Joanna Kopecka
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Giuseppe De Rosa
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy
| | - Thomas Mohr
- Center for Cancer Research and Comprehensive Cancer Center, Medical University Vienna, Vienna, Austria
| | - Alessandro Bertero
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino, Italy
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy
| | - Luisella Righi
- Pathology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Torino, Italy
| | - Silvia Novello
- Thoracic Oncology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Torino, Italy
| | | | - Federico Bussolino
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
- IRCCS Candiolo Cancer Institute, Candiolo, Italy
| | - Chiara Riganti
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.
- Molecular Biotechnology Center "Guido Tarone", University of Torino, Torino, Italy.
| |
Collapse
|
12
|
Dai M, Lin B, Li H, Wang Y, Wu M, Wei Y, Zeng W, Qu L, Cang C, Wang X. Lysosomal cation channel TRPML1 suppression sensitizes acute myeloid leukemia cells to chemotherapeutics by inhibiting autophagy. Mol Cell Biochem 2024:10.1007/s11010-024-05054-5. [PMID: 38951379 DOI: 10.1007/s11010-024-05054-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 06/12/2024] [Indexed: 07/03/2024]
Abstract
Despite the implementation of novel therapeutic regimens and extensive research efforts, chemoresistance remains a formidable challenge in the treatment of acute myeloid leukemia (AML). Notably, the involvement of lysosomes in chemoresistance has sparked interest in developing lysosome-targeted therapies to sensitize tumor cells to currently approved chemotherapy or as innovative pharmacological approaches. Moreover, as ion channels on the lysosomal membrane are critical regulators of lysosomal function, they present potential as novel targets for enhancing chemosensitivity. Here, we discovered that the expression of a lysosomal cation channel, namely transient receptor potential mucolipin 1 (TRPML1), was elevated in AML cells. Inhibiting TRPML1 individually does not impact the proliferation and apoptosis of AML cells. Importantly, inhibition of TRPML1 demonstrated the potential to modulate the sensitivity of AML cells to chemotherapeutic agents. Exploration of the underlying mechanisms revealed that suppression of TRPML1 impaired autophagy while concurrently increasing the production of reactive oxygen species (ROS) and ROS-mediated lipid peroxidation (Lipid-ROS) in AML cells. Finally, the knockdown of TRPML1 significantly reduced OCI-AML3 tumor growth following chemotherapy in a mouse model of human leukemia. In summary, targeting TRPML1 represents a promising approach for combination therapy aimed at enhancing chemosensitivity in treating AML.
Collapse
Affiliation(s)
- Meifang Dai
- Department of Hematology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
| | - Bingqian Lin
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Hao Li
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Youming Wang
- Department of Hematology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Miaomiao Wu
- Department of Hematology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yanan Wei
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Wenping Zeng
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China
| | - Lili Qu
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, Anhui, China.
| | - Chunlei Cang
- CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China.
| | - Xingbing Wang
- Department of Hematology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
13
|
Azbazdar Y, Sosa EA, Monka J, Kurmangaliyev YZ, Tejeda-Muñoz N. Interactions between genistein and Wnt pathway in colon adenocarcinoma and early embryos. Heliyon 2024; 10:e32243. [PMID: 38947477 PMCID: PMC11214441 DOI: 10.1016/j.heliyon.2024.e32243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/30/2024] [Accepted: 05/30/2024] [Indexed: 07/02/2024] Open
Abstract
The Wnt signaling pathway is one of the most ancient and pivotal signaling cascades, governing diverse processes in development and cancer regulation. Within the realm of cancer treatment, genistein emerges as a promising candidate due to its multifaceted modulation of various signaling pathways, including the Wnt pathway. Despite promising preclinical studies, the precise mechanisms underlying genistein's therapeutic effects via Wnt modulation remain elusive. In this study, we unveil novel insights into the therapeutic mechanisms of genistein by elucidating its inhibitory effects on Wnt signaling through macropinocytosis. Additionally, we demonstrate its capability to curtail cell growth, proliferation, and lysosomal activity in the SW480 colon adenocarcinoma cell model. Furthermore, our investigation extends to the embryonic context, where genistein influences gene regulatory networks governed by endogenous Wnt pathways. Our findings shed light on the intricate interplay between genistein, Wnt signaling, membrane trafficking, and gene regulation, paving the way for further exploration of genistein's therapeutic potential in cancer treatment strategies.
Collapse
Affiliation(s)
- Yagmur Azbazdar
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
| | - Eric A. Sosa
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Julia Monka
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
| | | | - Nydia Tejeda-Muñoz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
14
|
Zhang P, Yue L, Leng Q, Chang C, Gan C, Ye T, Cao D. Targeting FGFR for cancer therapy. J Hematol Oncol 2024; 17:39. [PMID: 38831455 PMCID: PMC11149307 DOI: 10.1186/s13045-024-01558-1] [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: 03/11/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
The FGFR signaling pathway is integral to cellular activities, including proliferation, differentiation, and survival. Dysregulation of this pathway is implicated in numerous human cancers, positioning FGFR as a prominent therapeutic target. Here, we conduct a comprehensive review of the function, signaling pathways and abnormal alterations of FGFR, as well as its role in tumorigenesis and development. Additionally, we provide an in-depth analysis of pivotal phase 2 and 3 clinical trials evaluating the performance and safety of FGFR inhibitors in oncology, thereby shedding light on the current state of clinical research in this field. Then, we highlight four drugs that have been approved for marketing by the FDA, offering insights into their molecular mechanisms and clinical achievements. Our discussion encompasses the intricate landscape of FGFR-driven tumorigenesis, current techniques for pinpointing FGFR anomalies, and clinical experiences with FGFR inhibitor regimens. Furthermore, we discuss the inherent challenges of targeting the FGFR pathway, encompassing resistance mechanisms such as activation by gatekeeper mutations, alternative pathways, and potential adverse reactions. By synthesizing the current evidence, we underscore the potential of FGFR-centric therapies to enhance patient prognosis, while emphasizing the imperative need for continued research to surmount resistance and optimize treatment modalities.
Collapse
Affiliation(s)
- Pei Zhang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Lin Yue
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - QingQing Leng
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Chen Chang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Cailing Gan
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tinghong Ye
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Dan Cao
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
15
|
Cerda‐Troncoso C, Grünenwald F, Arias‐Muñoz E, Cavieres VA, Caceres‐Verschae A, Hernández S, Gaete‐Ramírez B, Álvarez‐Astudillo F, Acuña RA, Ostrowski M, Burgos PV, Varas‐Godoy M. Chemo-small extracellular vesicles released in cisplatin-resistance ovarian cancer cells are regulated by the lysosomal function. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e157. [PMID: 38947172 PMCID: PMC11212338 DOI: 10.1002/jex2.157] [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: 06/26/2023] [Revised: 04/03/2024] [Accepted: 04/29/2024] [Indexed: 07/02/2024]
Abstract
Chemoresistance is a common problem in ovarian cancer (OvCa) treatment, where resistant cells, in response to chemotherapy, secrete small extracellular vesicles (sEVs), known as chemo-sEVs, that transfer resistance to recipient cells. sEVs are formed as intraluminal vesicles (ILVs) within multivesicular endosomes (MVEs), whose trafficking is regulated by Ras-associated binding (RAB) GTPases that mediate sEVs secretion or lysosomal degradation. A decrease in lysosomal function can promote sEVs secretion, but the relationship between MVEs trafficking pathways and sEVs secretion in OvCa chemoresistance is unclear. Here, we show that A2780cis cisplatin (CCDP) resistant OvCa cells had an increased number of MVEs and ILVs structures, higher levels of Endosomal Sorting Complex Required for Transport (ESCRTs) machinery components, and RAB27A compared to A2780 CDDP-sensitive OvCa cells. CDDP promoted the secretion of chemo-sEVs in A2780cis cells, enriched in DNA damage response proteins. A2780cis cells exhibited poor lysosomal function with reduced levels of RAB7, essential in MVEs-Lysosomal trafficking. The silencing of RAB27A in A2780cis cells prevents the Chemo-EVs secretion, reduces its chemoresistance and restores lysosomal function and levels of RAB7, switching them into an A2780-like cellular phenotype. Enhancing lysosomal function with rapamycin reduced chemo-sEVs secretion. Our results suggest that adjusting the balance between secretory MVEs and lysosomal MVEs trafficking could be a promising strategy for overcoming CDDP chemoresistance in OvCa.
Collapse
Affiliation(s)
- Cristóbal Cerda‐Troncoso
- Organelle Phagy Lab, CEBICEMFacultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
- Cancer Cell Biology Lab, CEBICEM, Facultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
- Centro Ciencia & VidaFundación Ciencia & VidaSantiagoChile
| | - Felipe Grünenwald
- Cancer Cell Biology Lab, CEBICEM, Facultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
| | - Eloísa Arias‐Muñoz
- Organelle Phagy Lab, CEBICEMFacultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
| | - Viviana A. Cavieres
- Organelle Phagy Lab, CEBICEMFacultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
- Departamento de Ciencias Biológicas y Químicas, Facultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
| | - Albano Caceres‐Verschae
- Cancer Cell Biology Lab, CEBICEM, Facultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
| | - Sergio Hernández
- Organelle Phagy Lab, CEBICEMFacultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
| | - Belén Gaete‐Ramírez
- Cancer Cell Biology Lab, CEBICEM, Facultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
| | | | - Rodrigo A. Acuña
- Centro de Medicina Regenerativa, Facultad de MedicinaClínica Alemana Universidad del DesarrolloSantiagoChile
| | - Matias Ostrowski
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS)Universidad de Buenos Aires (UBA)Buenos AiresArgentina
| | - Patricia V. Burgos
- Organelle Phagy Lab, CEBICEMFacultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
- Centro Ciencia & VidaFundación Ciencia & VidaSantiagoChile
| | - Manuel Varas‐Godoy
- Cancer Cell Biology Lab, CEBICEM, Facultad de Medicina y CienciaUniversidad San SebastiánSantiagoChile
- Centro Ciencia & VidaFundación Ciencia & VidaSantiagoChile
- Advanced Center for Chronic DiseasesSantiagoChile
| |
Collapse
|
16
|
Wang KN, Zhou K, Zhong NN, Cao LM, Li ZZ, Xiao Y, Wang GR, Huo FY, Zhou JJ, Liu B, Bu LL. Enhancing cancer therapy: The role of drug delivery systems in STAT3 inhibitor efficacy and safety. Life Sci 2024; 346:122635. [PMID: 38615745 DOI: 10.1016/j.lfs.2024.122635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/14/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
The signal transducer and activator of transcription 3 (STAT3), a member of the STAT family, resides in the nucleus to regulate genes essential for vital cellular functions, including survival, proliferation, self-renewal, angiogenesis, and immune response. However, continuous STAT3 activation in tumor cells promotes their initiation, progression, and metastasis, rendering STAT3 pathway inhibitors a promising avenue for cancer therapy. Nonetheless, these inhibitors frequently encounter challenges such as cytotoxicity and suboptimal biocompatibility in clinical trials. A viable strategy to mitigate these issues involves delivering STAT3 inhibitors via drug delivery systems (DDSs). This review delineates the regulatory mechanisms of the STAT3 signaling pathway and its association with cancer. It offers a comprehensive overview of the current application of DDSs for anti-STAT3 inhibitors and investigates the role of DDSs in cancer treatment. The conclusion posits that DDSs for anti-STAT3 inhibitors exhibit enhanced efficacy and reduced adverse effects in tumor therapy compared to anti-STAT3 inhibitors alone. This paper aims to provide an outline of the ongoing research and future prospects of DDSs for STAT3 inhibitors. Additionally, it presents our insights on the merits and future outlook of DDSs in cancer treatment.
Collapse
Affiliation(s)
- Kang-Ning Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Kan Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Fang-Yi Huo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jun-Jie Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial, Anyang Sixth People's Hospital, Anyang 45500, China.
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
17
|
Dunnington EL, Wong BS, Fu D. Innovative Approaches for Drug Discovery: Quantifying Drug Distribution and Response with Raman Imaging. Anal Chem 2024; 96:7926-7944. [PMID: 38625100 PMCID: PMC11108735 DOI: 10.1021/acs.analchem.4c01413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Affiliation(s)
| | | | - Dan Fu
- Department of Chemistry, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
18
|
Gomes LS, Costa ÉDO, Duarte TG, Charret TS, Castiglione RC, Simões RL, Pascoal VDB, Döring TH, da Silva FDC, Ferreira VF, S. de Oliveira A, Pascoal ACRF, Cruz AL, Nascimento V. New Chalcogen-Functionalized Naphthoquinones: Design, Synthesis, and Evaluation, In Vitro and In Silico, against Squamous Cell Carcinoma. ACS OMEGA 2024; 9:21948-21963. [PMID: 38799368 PMCID: PMC11112715 DOI: 10.1021/acsomega.3c10134] [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: 12/18/2023] [Revised: 04/24/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024]
Abstract
Due to the growth in the number of patients and the complexity involved in anticancer therapies, new therapeutic approaches are urgent and necessary. In this context, compounds containing the selenium atom can be employed in developing new medicines due to their potential therapeutic efficacy and unique modes of action. Furthermore, tellurium, a previously unknown element, has emerged as a promising possibility in chalcogen-containing compounds. In this study, 13 target compounds (9a-i, 10a-c, and 11) were effectively synthesized as potential anticancer agents, employing a CuI-catalyzed Csp-chalcogen bond formation procedure. The developed methodology yielded excellent results, ranging from 30 to 85%, and the compounds were carefully characterized. Eight of these compounds showed promise as potential therapeutic drugs due to their high yields and remarkable selectivity against SCC-9 cells (squamous cell carcinoma). Compound 10a, in particular, demonstrated exceptional selectivity, making it an excellent choice for cancer cell targeting while sparing healthy cells. Furthermore, complementing in silico and molecular docking studies shed light on their physical features and putative modes of action. This research highlights the potential of these compounds in anticancer treatments and lays the way for future drug development efforts.
Collapse
Affiliation(s)
- Luana
da Silva Gomes
- SupraSelen
Laboratory, Department of Organic Chemistry, Institute of Chemistry, Federal University Fluminense, Campus of Valonguinho, Niterói-RJ 24020-141, Brazil
| | - Érica de Oliveira Costa
- SupraSelen
Laboratory, Department of Organic Chemistry, Institute of Chemistry, Federal University Fluminense, Campus of Valonguinho, Niterói-RJ 24020-141, Brazil
| | - Thuany G. Duarte
- SupraSelen
Laboratory, Department of Organic Chemistry, Institute of Chemistry, Federal University Fluminense, Campus of Valonguinho, Niterói-RJ 24020-141, Brazil
| | - Thiago S. Charret
- Research
Laboratory of Natural Products and Bioactive Molecules, Nova Friburgo
Health Institute, Fluminense Federal University
(ISNF-UFF), Nova Friburgo-RJ 28625-650, Brazil
| | - Raquel C. Castiglione
- Laboratory
for Clinical and Experimental Research on Vascular Biology, Biomedical
Center, State University of Rio de Janeiro, Rio de Janeiro-RJ 20550-900, Brazil
| | - Rafael L. Simões
- Laboratory
of Molecular and Cellular Pharmacology, Roberto Alcântara Gomes
Biology Institute, State University of Rio
de Janeiro, Rio de Janeiro 20551-030, Brazil
| | - Vinicius D. B. Pascoal
- Research
Laboratory of Natural Products and Bioactive Molecules, Nova Friburgo
Health Institute, Fluminense Federal University
(ISNF-UFF), Nova Friburgo-RJ 28625-650, Brazil
| | - Thiago H. Döring
- Department
of Exact Sciences and Education, Federal
University of Santa Catarina, Campus Blumenau, Blumenau-SC, 89036-256, Brazil
| | - Fernando de C. da Silva
- Applied Organic
Synthesis Laboratory (LabSOA), Institute of Chemistry, Universidade Federal Fluminense, Niterói-RJ 24020-141, Brazil
| | - Vitor F. Ferreira
- Department
of Exact Sciences and Education, Federal
University of Santa Catarina, Campus Blumenau, Blumenau-SC 89036-256, Brazil
| | - Aldo S. de Oliveira
- Department
of Exact Sciences and Education, Federal
University of Santa Catarina, Campus Blumenau, Blumenau-SC, 89036-256, Brazil
| | - Aislan C. R. F. Pascoal
- Research
Laboratory of Natural Products and Bioactive Molecules, Nova Friburgo
Health Institute, Fluminense Federal University
(ISNF-UFF), Nova Friburgo-RJ 28625-650, Brazil
| | - André L.
S. Cruz
- Physiopathology
Laboratory, Institute of Medical Sciences, Multidisciplinary Center
UFRJ, Federal University of Rio De Janeiro
(UFRJ), Macaé-RJ 27930-560, Brazil
| | - Vanessa Nascimento
- SupraSelen
Laboratory, Department of Organic Chemistry, Institute of Chemistry, Federal University Fluminense, Campus of Valonguinho, Niterói-RJ 24020-141, Brazil
| |
Collapse
|
19
|
Tejeda-Muñoz N, Azbazdar Y, Sosa EA, Monka J, Wei PS, Binder G, Mei KC, Kurmangaliyev YZ, De Robertis EM. Na,K-ATPase activity promotes macropinocytosis in colon cancer via Wnt signaling. Biol Open 2024; 13:bio060269. [PMID: 38713004 PMCID: PMC11139033 DOI: 10.1242/bio.060269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/18/2024] [Indexed: 05/08/2024] Open
Abstract
Recent research has shown that membrane trafficking plays an important role in canonical Wnt signaling through sequestration of the β-catenin destruction complex inside multivesicular bodies (MVBs) and lysosomes. In this study, we introduce Ouabain, an inhibitor of the Na,K-ATPase pump that establishes electric potentials across membranes, as a potent inhibitor of Wnt signaling. We find that Na,K-ATPase levels are elevated in advanced colon carcinoma, that this enzyme is elevated in cancer cells with constitutively activated Wnt pathway and is activated by GSK3 inhibitors that increase macropinocytosis. Ouabain blocks macropinocytosis, which is an essential step in Wnt signaling, probably explaining the strong effects of Ouabain on this pathway. In Xenopus embryos, brief Ouabain treatment at the 32-cell stage, critical for the earliest Wnt signal in development-inhibited brains, could be reversed by treatment with Lithium chloride, a Wnt mimic. Inhibiting membrane trafficking may provide a way of targeting Wnt-driven cancers.
Collapse
Affiliation(s)
- Nydia Tejeda-Muñoz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles 90095-1662, USA
- Department of Oncology Science, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- OU Health Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Yagmur Azbazdar
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles 90095-1662, USA
| | - Eric A. Sosa
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Julia Monka
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles 90095-1662, USA
| | - Pu-Sheng Wei
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Binghamton, Johnson City, NY 13790, USA
| | - Grace Binder
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles 90095-1662, USA
| | - Kuo-Ching Mei
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Binghamton, Johnson City, NY 13790, USA
| | | | - Edward M. De Robertis
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles 90095-1662, USA
| |
Collapse
|
20
|
Wan P, Zhong L, Yu L, Shen C, Shao X, Chen S, Zhou Z, Wang M, Zhang H, Liu B. Lysosome-related genes predict acute myeloid leukemia prognosis and response to immunotherapy. Front Immunol 2024; 15:1384633. [PMID: 38799454 PMCID: PMC11117069 DOI: 10.3389/fimmu.2024.1384633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
Background Acute myeloid leukemia (AML) is a highly aggressive and pathogenic hematologic malignancy with consistently high mortality. Lysosomes are organelles involved in cell growth and metabolism that fuse to form specialized Auer rods in AML, and their role in AML has not been elucidated. This study aimed to identify AML subtypes centered on lysosome-related genes and to construct a prognostic model to guide individualized treatment of AML. Methods Gene expression data and clinical data from AML patients were downloaded from two high-throughput sequencing platforms. The 191 lysosomal signature genes were obtained from the database MsigDB. Lysosomal clusters were identified by unsupervised consensus clustering. The differences in molecular expression, biological processes, and the immune microenvironment among lysosomal clusters were subsequently analyzed. Based on the molecular expression differences between lysosomal clusters, lysosomal-related genes affecting AML prognosis were screened by univariate cox regression and multivariate cox regression analyses. Algorithms for LASSO regression analyses were employed to construct prognostic models. The risk factor distribution, KM survival curve, was applied to evaluate the survival distribution of the model. Time-dependent ROC curves, nomograms and calibration curves were used to evaluate the predictive performance of the prognostic models. TIDE scores and drug sensitivity analyses were used to explore the implication of the model for AML treatment. Results Our study identified two lysosomal clusters, cluster1 has longer survival time and stronger immune infiltration compared to cluster2. The differences in biological processes between the two lysosomal clusters are mainly manifested in the lysosomes, vesicles, immune cell function, and apoptosis. The prognostic model consisting of six prognosis-related genes was constructed. The prognostic model showed good predictive performance in all three data sets. Patients in the low-risk group survived significantly longer than those in the high-risk group and had higher immune infiltration and stronger response to immunotherapy. Patients in the high-risk group showed greater sensitivity to cytarabine, imatinib, and bortezomib, but lower sensitivity to ATRA compared to low -risk patients. Conclusion Our prognostic model based on lysosome-related genes can effectively predict the prognosis of AML patients and provide reference evidence for individualized immunotherapy and pharmacological chemotherapy for AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/diagnosis
- Lysosomes/metabolism
- Prognosis
- Female
- Male
- Immunotherapy/methods
- Biomarkers, Tumor/genetics
- Middle Aged
- Gene Expression Profiling
- Adult
- Nomograms
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
- Aged
- Gene Expression Regulation, Leukemic
- Transcriptome
Collapse
Affiliation(s)
- Peng Wan
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Liang Zhong
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Lihua Yu
- Clinical Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Chenlan Shen
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Xin Shao
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Shuyu Chen
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Ziwei Zhou
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Meng Wang
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Hongyan Zhang
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
| | - Beizhong Liu
- Central Laboratory of Yongchuan Hospital, Chongqing Medical University, Chongqing, China
- Key Laboratory of Laboratory Medical Diagnostics, Ministry of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Offensperger F, Tin G, Duran-Frigola M, Hahn E, Dobner S, Ende CWA, Strohbach JW, Rukavina A, Brennsteiner V, Ogilvie K, Marella N, Kladnik K, Ciuffa R, Majmudar JD, Field SD, Bensimon A, Ferrari L, Ferrada E, Ng A, Zhang Z, Degliesposti G, Boeszoermenyi A, Martens S, Stanton R, Müller AC, Hannich JT, Hepworth D, Superti-Furga G, Kubicek S, Schenone M, Winter GE. Large-scale chemoproteomics expedites ligand discovery and predicts ligand behavior in cells. Science 2024; 384:eadk5864. [PMID: 38662832 DOI: 10.1126/science.adk5864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 03/22/2024] [Indexed: 05/04/2024]
Abstract
Chemical modulation of proteins enables a mechanistic understanding of biology and represents the foundation of most therapeutics. However, despite decades of research, 80% of the human proteome lacks functional ligands. Chemical proteomics has advanced fragment-based ligand discovery toward cellular systems, but throughput limitations have stymied the scalable identification of fragment-protein interactions. We report proteome-wide maps of protein-binding propensity for 407 structurally diverse small-molecule fragments. We verified that identified interactions can be advanced to active chemical probes of E3 ubiquitin ligases, transporters, and kinases. Integrating machine learning binary classifiers further enabled interpretable predictions of fragment behavior in cells. The resulting resource of fragment-protein interactions and predictive models will help to elucidate principles of molecular recognition and expedite ligand discovery efforts for hitherto undrugged proteins.
Collapse
Affiliation(s)
- Fabian Offensperger
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Gary Tin
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Miquel Duran-Frigola
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
- Ersilia Open Source Initiative, Cambridge CB1 3DE, UK
| | - Elisa Hahn
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Sarah Dobner
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | | | | | - Andrea Rukavina
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Vincenth Brennsteiner
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Kevin Ogilvie
- Medicine Design, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Nara Marella
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Katharina Kladnik
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Rodolfo Ciuffa
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | | | | | - Ariel Bensimon
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Luca Ferrari
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna Biocenter 5, 1030 Vienna, Austria
- University of Vienna, Max Perutz Labs, Vienna Biocenter 5, 1030 Vienna, Austria
| | - Evandro Ferrada
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Amanda Ng
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Zhechun Zhang
- Molecular Informatics, Machine Learning and Computational Sciences, Early Clinical Development, Pfizer, Cambridge, MA 02139, USA
| | - Gianluca Degliesposti
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Andras Boeszoermenyi
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Sascha Martens
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna Biocenter 5, 1030 Vienna, Austria
- University of Vienna, Max Perutz Labs, Vienna Biocenter 5, 1030 Vienna, Austria
| | - Robert Stanton
- Molecular Informatics, Machine Learning and Computational Sciences, Early Clinical Development, Pfizer, Cambridge, MA 02139, USA
| | - André C Müller
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - J Thomas Hannich
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | | | - Giulio Superti-Furga
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
- Center for Physiology and Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
| | - Stefan Kubicek
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | | | - Georg E Winter
- CeMM, Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| |
Collapse
|
22
|
Lu XX, Xue C, Dong JH, Zhang YZ, Gao F. Nanoplatform-based strategies for enhancing the lethality of current antitumor PDT. J Mater Chem B 2024; 12:3209-3225. [PMID: 38497405 DOI: 10.1039/d4tb00008k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Photodynamic therapy (PDT) exhibits great application prospects in future clinical oncology due to its spatiotemporal controllability and good biosafety. However, the antitumor efficacy of PDT is seriously hindered by many factors, including tumor hypoxia, limited light penetration ability, and strong defense mechanisms of tumors. Considering that it is difficult to completely solve the first two problems, enhancing the lethality of antitumor PDT has become a good idea to extend its clinical application. Herein, we summarize the nanoplatform-involved strategies to effectively amplify the tumoricidal capability of current PDT and then discuss the present bottlenecks and prospects of the nanoplatform-based PDT sensitization strategies in tumor therapy. We hope this review will provide some references for others to design high-performance PDT nanoplatforms for tumor therapy.
Collapse
Affiliation(s)
- Xin-Xin Lu
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| | - Chun Xue
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| | - Jian-Hui Dong
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| | - Yi-Zhou Zhang
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| | - Fan Gao
- Institute of Advanced Materials and Flexible Electronics (IAMFE), School of Chemistry and Materials Science, Nanjing University of Information Science & Technology, Nanjing, 210044, China.
| |
Collapse
|
23
|
Fu XY, Yin H, Chen XT, Yao JF, Ma YN, Song M, Xu H, Yu QY, Du SS, Qi YK, Wang KW. Three Rounds of Stability-Guided Optimization and Systematical Evaluation of Oncolytic Peptide LTX-315. J Med Chem 2024; 67:3885-3908. [PMID: 38278140 DOI: 10.1021/acs.jmedchem.3c02232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2024]
Abstract
Oncolytic peptides represent promising novel candidates for anticancer treatments. In our efforts to develop oncolytic peptides possessing both high protease stability and durable anticancer efficiency, three rounds of optimization were conducted on the first-in-class oncolytic peptide LTX-315. The robust synthetic method, in vitro and in vivo anticancer activity, and anticancer mechanism were investigated. The D-type peptides represented by FXY-12 possessed significantly improved proteolytic stability and sustained anticancer efficiency. Strikingly, the novel hybrid peptide FXY-30, containing one FXY-12 and two camptothecin moieties, exhibited the most potent in vitro and in vivo anticancer activities. The mechanism explorations indicated that FXY-30 exhibited rapid membranolytic effects and induced severe DNA double-strand breaks to trigger cell apoptosis. Collectively, this study not only established robust strategies to improve the stability and anticancer potential of oncolytic peptides but also provided valuable references for the future development of D-type peptides-based hybrid anticancer chemotherapeutics.
Collapse
Affiliation(s)
- Xing-Yan Fu
- School of Pharmacy, Qingdao University Medical College, Qingdao University, #1 Ningde Road, Qingdao 266073, China
- Institute of Innovative Drugs, Qingdao University, #38 Dengzhou Road, Qingdao 266021, China
| | - Hao Yin
- School of Pharmacy, Qingdao University Medical College, Qingdao University, #1 Ningde Road, Qingdao 266073, China
- Institute of Innovative Drugs, Qingdao University, #38 Dengzhou Road, Qingdao 266021, China
| | - Xi-Tong Chen
- School of Pharmacy, Qingdao University Medical College, Qingdao University, #1 Ningde Road, Qingdao 266073, China
| | - Jing-Fang Yao
- School of Pharmacy, Qingdao University Medical College, Qingdao University, #1 Ningde Road, Qingdao 266073, China
| | - Yan-Nan Ma
- School of Pharmacy, Qingdao University Medical College, Qingdao University, #1 Ningde Road, Qingdao 266073, China
| | - Min Song
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Huan Xu
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Qian-Yao Yu
- School of Pharmacy, Qingdao University Medical College, Qingdao University, #1 Ningde Road, Qingdao 266073, China
| | - Shan-Shan Du
- School of Pharmacy, Qingdao University Medical College, Qingdao University, #1 Ningde Road, Qingdao 266073, China
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yun-Kun Qi
- School of Pharmacy, Qingdao University Medical College, Qingdao University, #1 Ningde Road, Qingdao 266073, China
- Institute of Innovative Drugs, Qingdao University, #38 Dengzhou Road, Qingdao 266021, China
| | - Ke-Wei Wang
- School of Pharmacy, Qingdao University Medical College, Qingdao University, #1 Ningde Road, Qingdao 266073, China
- Institute of Innovative Drugs, Qingdao University, #38 Dengzhou Road, Qingdao 266021, China
| |
Collapse
|
24
|
Gagliardi S, Mitruccio M, Di Corato R, Romano R, Aloisi A, Rinaldi R, Alifano P, Guerra F, Bucci C. Defects of mitochondria-lysosomes communication induce secretion of mitochondria-derived vesicles and drive chemoresistance in ovarian cancer cells. Cell Commun Signal 2024; 22:165. [PMID: 38448982 PMCID: PMC10916030 DOI: 10.1186/s12964-024-01507-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 01/31/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Among the mechanisms of mitochondrial quality control (MQC), generation of mitochondria-derived vesicles (MDVs) is a process to avoid complete failure of mitochondria determining lysosomal degradation of mitochondrial damaged proteins. In this context, RAB7, a late endocytic small GTPase, controls delivery of MDVs to late endosomes for subsequent lysosomal degradation. We previously demonstrated that RAB7 has a pivotal role in response to cisplatin (CDDP) regulating resistance to the drug by extracellular vesicle (EVs) secretion. METHODS Western blot and immunofluorescence analysis were used to analyze structure and function of endosomes and lysosomes in CDDP chemosensitive and chemoresistant ovarian cancer cell lines. EVs were purified from chemosensitive and chemoresistant cells by ultracentrifugation or immunoisolation to analyze their mitochondrial DNA and protein content. Treatment with cyanide m-chlorophenylhydrazone (CCCP) and RAB7 modulation were used, respectively, to understand the role of mitochondrial and late endosomal/lysosomal alterations on MDV secretion. Using conditioned media from chemoresistant cells the effect of MDVs on the viability after CDDP treatment was determined. Seahorse assays and immunofluorescence analysis were used to study the biochemical role of MDVs and the uptake and intracellular localization of MDVs, respectively. RESULTS We observed that CDDP-chemoresistant cells are characterized by increased MDV secretion, impairment of late endocytic traffic, RAB7 downregulation, an increase of RAB7 in EVs, compared to chemosensitive cells, and downregulation of the TFEB-mTOR pathway overseeing lysosomal and mitochondrial biogenesis and turnover. We established that MDVs can be secreted rather than delivered to lysosomes and are able to deliver CDDP outside the cells. We showed increased secretion of MDVs by chemoresistant cells ultimately caused by the extrusion of RAB7 in EVs, resulting in a dramatic drop in its intracellular content, as a novel mechanism to regulate RAB7 levels. We demonstrated that MDVs purified from chemoresistant cells induce chemoresistance in RAB7-modulated process, and, after uptake from recipient cells, MDVs localize to mitochondria and slow down mitochondrial activity. CONCLUSIONS Dysfunctional MQC in chemoresistant cells determines a block in lysosomal degradation of MDVs and their consequent secretion, suggesting that MQC is not able to eliminate damaged mitochondria whose components are secreted becoming effectors and potential markers of chemoresistance.
Collapse
Affiliation(s)
- Sinforosa Gagliardi
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Marco Mitruccio
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Riccardo Di Corato
- Institute for Microelectronics and Microsystems (IMM), CNR, Via Monteroni, Lecce, 73100, Italy
- Center for Biomolecular Nanotechnologies, Istituto Italiano di Tecnologia, Arnesano, 73010, Italy
| | - Roberta Romano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
- Department of Experimental Medicine, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Alessandra Aloisi
- Institute for Microelectronics and Microsystems (IMM), CNR, Via Monteroni, Lecce, 73100, Italy
| | - Rosaria Rinaldi
- Department of Mathematics and Physics "E. De Giorgi", University of Salento, Via Monteroni, Lecce, 73100, Italy
- Scuola Superiore ISUFI, University of Salento, Via Monteroni, University Campus, Lecce, 73100, Italy
| | - Pietro Alifano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
- Department of Experimental Medicine, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy.
- Department of Experimental Medicine, University of Salento, Via Provinciale Lecce-Monteroni n. 165, Lecce, 73100, Italy.
| |
Collapse
|
25
|
Eriksson I, Öllinger K. Lysosomes in Cancer-At the Crossroad of Good and Evil. Cells 2024; 13:459. [PMID: 38474423 DOI: 10.3390/cells13050459] [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: 12/21/2023] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Although it has been known for decades that lysosomes are central for degradation and recycling in the cell, their pivotal role as nutrient sensing signaling hubs has recently become of central interest. Since lysosomes are highly dynamic and in constant change regarding content and intracellular position, fusion/fission events allow communication between organelles in the cell, as well as cell-to-cell communication via exocytosis of lysosomal content and release of extracellular vesicles. Lysosomes also mediate different forms of regulated cell death by permeabilization of the lysosomal membrane and release of their content to the cytosol. In cancer cells, lysosomal biogenesis and autophagy are increased to support the increased metabolism and allow growth even under nutrient- and oxygen-poor conditions. Tumor cells also induce exocytosis of lysosomal content to the extracellular space to promote invasion and metastasis. However, due to the enhanced lysosomal function, cancer cells are often more susceptible to lysosomal membrane permeabilization, providing an alternative strategy to induce cell death. This review summarizes the current knowledge of cancer-associated alterations in lysosomal structure and function and illustrates how lysosomal exocytosis and release of extracellular vesicles affect disease progression. We focus on functional differences depending on lysosomal localization and the regulation of intracellular transport, and lastly provide insight how new therapeutic strategies can exploit the power of the lysosome and improve cancer treatment.
Collapse
Affiliation(s)
- Ida Eriksson
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden
| | - Karin Öllinger
- Division of Cell Biology, Department of Biomedical and Clinical Sciences, Linköping University, 58185 Linköping, Sweden
| |
Collapse
|
26
|
Chen Q, Fang C, Xia F, Wang Q, Li F, Ling D. Metal nanoparticles for cancer therapy: Precision targeting of DNA damage. Acta Pharm Sin B 2024; 14:1132-1149. [PMID: 38486992 PMCID: PMC10934341 DOI: 10.1016/j.apsb.2023.08.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/30/2023] [Accepted: 08/15/2023] [Indexed: 03/17/2024] Open
Abstract
Cancer, a complex and heterogeneous disease, arises from genomic instability. Currently, DNA damage-based cancer treatments, including radiotherapy and chemotherapy, are employed in clinical practice. However, the efficacy and safety of these therapies are constrained by various factors, limiting their ability to meet current clinical demands. Metal nanoparticles present promising avenues for enhancing each critical aspect of DNA damage-based cancer therapy. Their customizable physicochemical properties enable the development of targeted and personalized treatment platforms. In this review, we delve into the design principles and optimization strategies of metal nanoparticles. We shed light on the limitations of DNA damage-based therapy while highlighting the diverse strategies made possible by metal nanoparticles. These encompass targeted drug delivery, inhibition of DNA repair mechanisms, induction of cell death, and the cascading immune response. Moreover, we explore the pivotal role of physicochemical factors such as nanoparticle size, stimuli-responsiveness, and surface modification in shaping metal nanoparticle platforms. Finally, we present insights into the challenges and future directions of metal nanoparticles in advancing DNA damage-based cancer therapy, paving the way for novel treatment paradigms.
Collapse
Affiliation(s)
- Qian Chen
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Chunyan Fang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Fan Xia
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qiyue Wang
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
| | - Fangyuan Li
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, Hangzhou 310009, China
| | - Daishun Ling
- Institute of Pharmaceutics, Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Frontiers Science Center for Transformative Molecules, School of Chemistry and Chemical Engineering, National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
- World Laureates Association (WLA) Laboratories, Shanghai 201203, China
| |
Collapse
|
27
|
Tang H, Luo X, Shen X, Fan D, Rao J, Wan Y, Ma H, Guo X, Liu Z, Gao J. Lysosome-related biomarkers in preeclampsia and cancers: Machine learning and bioinformatics analysis. Comput Biol Med 2024; 171:108201. [PMID: 38428097 DOI: 10.1016/j.compbiomed.2024.108201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 01/21/2024] [Accepted: 02/18/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Lysosomes serve as regulatory hubs, and play a pivotal role in human diseases. However, the precise functions and mechanisms of action of lysosome-related genes remain unclear in preeclampsia and cancers. This study aimed to identify lysosome-related biomarkers in preeclampsia, and further explore the biomarkers shared between preeclampsia and cancers. MATERIALS AND METHODS We obtained GSE60438 and GSE75010 datasets from the Gene Expression Omnibus database, pre-procesed them and merged them into a training cohort. The limma package in R was used to identify the differentially expressed mRNAs between the preeclampsia and normal control groups. Differentially expressed lysosome-related genes were identified by intersecting the differentially expressed mRNAs and lysosome-related genes obtained from Gene Ontology and GSEA databases. Gene Ontology annotations and Kyoto Encyclopedia of Genes and Genomes enrichment analysis were performed using the DAVID database. The CIBERSORT method was used to analyze immune cell infiltration. Weighted gene co-expression analyses and three machine learning algorithm were used to identify lysosome-related diagnostic biomarkers. Lysosome-related diagnostic biomarkers were further validated in the testing cohort GSE25906. Nomogram diagnostic models for preeclampsia were constructed. In addition, pan-cancer analysis of lysosome-related diagnostic biomarkers were identified by was performed using the TIMER, Sangebox and TISIDB databases. Finally, the Drug-Gene Interaction, TheMarker and DSigDB Databases were used for drug-gene interactions analysis. RESULTS A total of 11 differentially expressed lysosome-related genes were identified between the preeclampsia and control groups. Three molecular clusters connected to lysosome were identified, and enrichment analysis demonstrated their strong relevance to the development and progression of preeclampsia. Immune infiltration analysis revealed significant immunity heterogeneity among different clusters. GBA, OCRL, TLR7 and HEXB were identified as lysosome-related diagnostic biomarkers with high AUC values, and validated in the testing cohort GSE25906. Nomogram, calibration curve, and decision curve analysis confirmed the accuracy of predicting the occurrence of preeclampsia based on OCRL and HEXB. Pan-cancer analysis showed that GBA, OCRL, TLR7 and HEXB were associated with the prognosis of patients with various tumors and tumor immune cell infiltration. Twelve drugs were identified as potential drugs for the treatment of preeclampsia and cancers. CONCLUSION This study identified GBA, OCRL, TLR7 and HEXB as potential lysosome-related diagnostic biomarkers shared between preeclampsia and cancers.
Collapse
Affiliation(s)
- Hai Tang
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, Guangdong, 510080, China; Foshan Institute of Fetal Medicine, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China; Department of Obstetrics, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Xin Luo
- Foshan Institute of Fetal Medicine, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China; Department of Obstetrics, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Xiuyin Shen
- Foshan Institute of Fetal Medicine, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China; Department of Obstetrics, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Dazhi Fan
- Foshan Institute of Fetal Medicine, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China; Department of Obstetrics, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Jiamin Rao
- Foshan Institute of Fetal Medicine, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China; Department of Obstetrics, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Yingchun Wan
- Foshan Institute of Fetal Medicine, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China; Department of Obstetrics, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Huiting Ma
- Foshan Institute of Fetal Medicine, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China; Department of Obstetrics, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Xiaoling Guo
- Foshan Institute of Fetal Medicine, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China; Department of Obstetrics, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Zhengping Liu
- Foshan Institute of Fetal Medicine, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China; Department of Obstetrics, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, 528000, China.
| | - Jie Gao
- Premarital Examination and Superior Examination Department, Jingzhou Gongan Maternal and Child Health Care Hospital, Jingzhou, Hubei, 434300, China.
| |
Collapse
|
28
|
To KKW, Huang Z, Zhang H, Ashby CR, Fu L. Utilizing non-coding RNA-mediated regulation of ATP binding cassette (ABC) transporters to overcome multidrug resistance to cancer chemotherapy. Drug Resist Updat 2024; 73:101058. [PMID: 38277757 DOI: 10.1016/j.drup.2024.101058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/27/2023] [Accepted: 01/16/2024] [Indexed: 01/28/2024]
Abstract
Multidrug resistance (MDR) is one of the primary factors that produces treatment failure in patients receiving cancer chemotherapy. MDR is a complex multifactorial phenomenon, characterized by a decrease or abrogation of the efficacy of a wide spectrum of anticancer drugs that are structurally and mechanistically distinct. The overexpression of the ATP-binding cassette (ABC) transporters, notably ABCG2 and ABCB1, are one of the primary mediators of MDR in cancer cells, which promotes the efflux of certain chemotherapeutic drugs from cancer cells, thereby decreasing or abolishing their therapeutic efficacy. A number of studies have suggested that non-coding RNAs (ncRNAs), particularly microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), play a pivotal role in mediating the upregulation of ABC transporters in certain MDR cancer cells. This review will provide updated information about the induction of ABC transporters due to the aberrant regulation of ncRNAs in cancer cells. We will also discuss the measurement and biological profile of circulating ncRNAs in various body fluids as potential biomarkers for predicting the response of cancer patients to chemotherapy. Sequence variations, such as alternative polyadenylation of mRNA and single nucleotide polymorphism (SNPs) at miRNA target sites, which may indicate the interaction of miRNA-mediated gene regulation with genetic variations to modulate the MDR phenotype, will be reviewed. Finally, we will highlight novel strategies that could be used to modulate ncRNAs and circumvent ABC transporter-mediated MDR.
Collapse
Affiliation(s)
- Kenneth K W To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region.
| | - Zoufang Huang
- Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Hang Zhang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, United States
| | - Liwu Fu
- State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine; Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
29
|
Khan SU, Fatima K, Aisha S, Malik F. Unveiling the mechanisms and challenges of cancer drug resistance. Cell Commun Signal 2024; 22:109. [PMID: 38347575 PMCID: PMC10860306 DOI: 10.1186/s12964-023-01302-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 08/30/2023] [Indexed: 02/15/2024] Open
Abstract
Cancer treatment faces many hurdles and resistance is one among them. Anti-cancer treatment strategies are evolving due to innate and acquired resistance capacity, governed by genetic, epigenetic, proteomic, metabolic, or microenvironmental cues that ultimately enable selected cancer cells to survive and progress under unfavorable conditions. Although the mechanism of drug resistance is being widely studied to generate new target-based drugs with better potency than existing ones. However, due to the broader flexibility in acquired drug resistance, advanced therapeutic options with better efficacy need to be explored. Combination therapy is an alternative with a better success rate though the risk of amplified side effects is commonplace. Moreover, recent groundbreaking precision immune therapy is one of the ways to overcome drug resistance and has revolutionized anticancer therapy to a greater extent with the only limitation of being individual-specific and needs further attention. This review will focus on the challenges and strategies opted by cancer cells to withstand the current therapies at the molecular level and also highlights the emerging therapeutic options -like immunological, and stem cell-based options that may prove to have better potential to challenge the existing problem of therapy resistance. Video Abstract.
Collapse
Affiliation(s)
- Sameer Ullah Khan
- Division of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Holcombe Blvd, Houston, TX, 77030, USA.
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| | - Kaneez Fatima
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Shariqa Aisha
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Fayaz Malik
- Division of Cancer Pharmacology, CSIR-Indian Institute of Integrative Medicine, Srinagar-190005, Jammu and Kashmir, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India.
| |
Collapse
|
30
|
Zhang H, Song J, Ward R, Han Y, Hunt A, Shriwas P, Steed A, Edwards C, Cao Y, Co M, Chen X. Diverse temporal and spatial mechanisms work, partially through Stanniocalcin-1, V-ATPase and senescence, to activate the extracellular ATP-mediated drug resistance in human cancer cells. Front Oncol 2024; 14:1276092. [PMID: 38380370 PMCID: PMC10876858 DOI: 10.3389/fonc.2024.1276092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 01/17/2024] [Indexed: 02/22/2024] Open
Abstract
Introduction Resistance to drug therapies is associated with a large majority of cancer-related deaths. ATP-binding cassette (ABC) transporter-mediated drug efflux, epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs), glutathione (GSH), senescence, and vacuole-type ATPase (V-ATPase) all contribute to the resistance. We recently showed that extracellular ATP (eATP) induces and regulates EMT, CSC formation, and ABC transporters in human cancer cells and tumors. eATP also consistently upregulates Stanniocalcin-1 (STC1), a gene that significantly contributes to EMT, CSC formation, and tumor growth. We also found that eATP enhances drug resistance in cancer cells through eATP internalization mediated by macropinocytosis, leading to an elevation of intracellular ATP (iATP) levels, induction of EMT, and CSC formation. However, these factors have never been systematically investigated in the context of eATP-induced drug resistance. Methods In this study, we hypothesized that eATP increases drug resistance via inducing ABC efflux, EMT, CSCs, STC1, and their accompanied processes such as GSH reducing activity, senescence, and V-ATPase. RNA sequencing, metabolomics, gene knockdown and knockout, and functional assays were performed to investigate these pathways and processes. Results and discussion Our study results showed that, in multiple human cancer lines, eATP induced genes involved in drug resistance, elevated ABC transporters' efflux activity of anticancer drugs; generated transcriptomic and metabolic profiles representing a drug resistant state; upregulated activities of GSH, senescence, and V-ATPase to promote drug resistance. Collectively, these newly found players shed light on the mechanisms of eATP-induced as well as STC1- and V-ATPase-mediated drug resistance and offer potential novel targets for combating drug resistance in cancers.
Collapse
Affiliation(s)
- Haiyun Zhang
- Department of Biological Science, Ohio University, Athens, OH, United States
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- The Program of Molecular and Cellular Biology, Ohio University, Athens, OH, United States
| | - Jingwen Song
- Department of Biological Science, Ohio University, Athens, OH, United States
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- The Program of Molecular and Cellular Biology, Ohio University, Athens, OH, United States
| | - Ryan Ward
- The Honor Tutorial College, Ohio University, Athens, OH, United States
| | - Yong Han
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
| | - Arabella Hunt
- The Honor Tutorial College, Ohio University, Athens, OH, United States
| | - Pratik Shriwas
- Department of Biological Science, Ohio University, Athens, OH, United States
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- The Program of Molecular and Cellular Biology, Ohio University, Athens, OH, United States
| | - Alexander Steed
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Cory Edwards
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Yanyang Cao
- Department of Biological Science, Ohio University, Athens, OH, United States
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- The Program of Molecular and Cellular Biology, Ohio University, Athens, OH, United States
| | - Milo Co
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| | - Xiaozhuo Chen
- Department of Biological Science, Ohio University, Athens, OH, United States
- The Edison Biotechnology Institute, Ohio University, Athens, OH, United States
- The Program of Molecular and Cellular Biology, Ohio University, Athens, OH, United States
- Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
- Department of Biomedical Science, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, United States
| |
Collapse
|
31
|
Wong BS, Dunnington EL, Wu R, Kim JI, Hu K, Ro TH, Fu D. Facilitated Transport of EGFR Inhibitors Plays an Important Role in Their Cellular Uptake. Anal Chem 2024; 96:1547-1555. [PMID: 38214696 PMCID: PMC11012238 DOI: 10.1021/acs.analchem.3c04242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Epidermal growth factor receptor (EGFR) is a transmembrane protein commonly targeted by tyrosine kinase inhibitors (TKIs) as a front-line therapy for patients with many cancers including nonsmall cell lung cancer (NSCLC). Effective treatment requires efficient intracellular drug uptake and target binding. However, despite the recent success in the development of new TKI drugs, the mechanisms of uptake for many TKIs are still poorly understood due to the difficulty in imaging and measuring nonfluorescent drug molecules at a subcellular resolution. It has previously been shown that weakly basic TKI drugs are sequestered in lysosomes. Leveraging this property, we apply hyperspectral stimulated Raman scattering imaging to directly visualize and quantify two Food and Drug Administration-approved EGFR inhibitor drugs (lapatinib and afatinib) inside living cells and the changes in their cellular uptake upon the addition of organic cation transporter inhibitors. These single-cell quantitative measurements provide new insight into the role of membrane transporters in the uptake of TKI drugs in living cells.
Collapse
Affiliation(s)
- Brian S Wong
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Erin L Dunnington
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Ruibing Wu
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Jonathan I Kim
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Kailun Hu
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Thomas H Ro
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| | - Dan Fu
- Department of Chemistry, University of Washington, Seattle, Washington 98195, United States
| |
Collapse
|
32
|
Jiang X, Huang K, Sun X, Li Y, Hua L, Liu F, Huang R, Du J, Zeng H. Hexamethylene amiloride synergizes with venetoclax to induce lysosome-dependent cell death in acute myeloid leukemia. iScience 2024; 27:108691. [PMID: 38205254 PMCID: PMC10776932 DOI: 10.1016/j.isci.2023.108691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/15/2023] [Accepted: 12/05/2023] [Indexed: 01/12/2024] Open
Abstract
Tumors maintain an alkaline intracellular environment to enable rapid growth. The proton exporter NHE1 participates in maintenance of this pH gradient. However, whether targeting NHE1 could inhibit the growth of tumor cells remains unknown. Here, we report that the NHE1 inhibitor Hexamethylene amiloride (HA) efficiently suppresses the growth of AML cell lines. Moreover, HA combined with venetoclax synergized to efficiently inhibit the growth of AML cells. Interestingly, lysosomes are the main contributors to the synergism of HA and venetoclax in inhibiting AML cells. Most importantly, the combination of HA and venetoclax also had prominent anti-leukemia effects in both xenograft models and bone marrow samples from AML patients. In summary, our results provide evidence that the NHE1 inhibitor HA or its combination with venetoclax efficiently inhibits the growth of AML in vitro and in vivo.
Collapse
Affiliation(s)
- Xinya Jiang
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Kexiu Huang
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
| | - Xiaofan Sun
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
| | - Yue Li
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
| | - Lei Hua
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
| | - Fangshu Liu
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
| | - Rui Huang
- Department of Hematology, Zhujiang Hospital of Southern Medical University, Guangzhou, P.R. China
| | - Juan Du
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
| | - Hui Zeng
- Department of Hematology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, China
| |
Collapse
|
33
|
Gao M, Deng H, Zhang Y, Wang H, Liu R, Hou W, Zhang W. Hyaluronan nanogel co-loaded with chloroquine to enhance intracellular cisplatin delivery through lysosomal permeabilization and lysophagy inhibition. Carbohydr Polym 2024; 323:121415. [PMID: 37940248 DOI: 10.1016/j.carbpol.2023.121415] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 11/10/2023]
Abstract
Hyaluronan (HA) has been widely used to construct nanocarriers for cancer-targeted drug delivery, due to its excellent biocompatibility and intrinsic affinity towards CD44 that is overexpressed in most cancer types. However, the HA-based nanocarriers are prone to trapping in lysosomes following the HA-mediated endocytosis, which limited the delivered drug to access its pharmacological action sites and subsequently compromised the therapeutic efficacy. To overcome this intracellular obstacle, here we demonstrated the co-loading of chloroquine (CQ) in HA nanogel could efficiently promote the intracellular delivery of cisplatin. The cisplatin coordination with HA generated the nanogel that could also co-encapsulate CQ (HA/Cis/CQ nanogel). Compared with cisplatin-loaded HA nanogel (HA/Cis), HA/Cis/CQ significantly promoted the lysosomal escape of cisplatin as well as enhanced tumor inhibition in the triple-negative breast cancer model. Mechanism studies suggested that co-delivery of CQ not only induced the lysosomal membrane permeabilization but also inhibited the lysophagy, which collectively contributed to the lysosomal instability and cisplatin escape. This HA/Cis/CQ nanogel elicited less toxicity compared with the combination of free Cis and CQ, thus suggesting a promising HA nanocarrier to boost the cisplatin delivery towards cancer-targeted therapy.
Collapse
Affiliation(s)
- Menghan Gao
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Hong Deng
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Yiyi Zhang
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Huimin Wang
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Runmeng Liu
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Wei Hou
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Weiqi Zhang
- State Key Laboratory of Complex, Severe, and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China.
| |
Collapse
|
34
|
Pandey G, Phatale V, Khairnar P, Kolipaka T, Shah S, Famta P, Jain N, Srinivasarao DA, Rajinikanth PS, Raghuvanshi RS, Srivastava S. Supramolecular self-assembled peptide-engineered nanofibers: A propitious proposition for cancer therapy. Int J Biol Macromol 2024; 256:128452. [PMID: 38042321 DOI: 10.1016/j.ijbiomac.2023.128452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/04/2023]
Abstract
Cancer is a devastating disease that causes a substantial number of deaths worldwide. Current therapeutic interventions for cancer include chemotherapy, radiation therapy, or surgery. These conventional therapeutic approaches are associated with disadvantages such as multidrug resistance, destruction of healthy tissues, and tissue toxicity. Therefore, there is a paradigm shift in cancer management wherein nanomedicine-based novel therapeutic interventions are being explored to overcome the aforementioned disadvantages. Supramolecular self-assembled peptide nanofibers are emerging drug delivery vehicles that have gained much attention in cancer management owing to their biocompatibility, biodegradability, biomimetic property, stimuli-responsiveness, transformability, and inherent therapeutic property. Supramolecules form well-organized structures via non-covalent linkages, the intricate molecular arrangement helps to improve tissue permeation, pharmacokinetic profile and chemical stability of therapeutic agents while enabling targeted delivery and allowing efficient tumor imaging. In this review, we present fundamental aspects of peptide-based self-assembled nanofiber fabrication their applications in monotherapy/combinatorial chemo- and/or immuno-therapy to overcome multi-drug resistance. The role of self-assembled structures in targeted/stimuli-responsive (pH, enzyme and photo-responsive) drug delivery has been discussed along with the case studies. Further, recent advancements in peptide nanofibers in cancer diagnosis, imaging, gene therapy, and immune therapy along with regulatory obstacles towards clinical translation have been deliberated.
Collapse
Affiliation(s)
- Giriraj Pandey
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Vivek Phatale
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pooja Khairnar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Tejaswini Kolipaka
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Saurabh Shah
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Paras Famta
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Naitik Jain
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Dadi A Srinivasarao
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Rajeev Singh Raghuvanshi
- Central Drugs Standard Control Organization (CDSCO), Directorate General of Health Services, Ministry of Health & Family Welfare, Government of India, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India.
| |
Collapse
|
35
|
Shirbhate E, Singh V, Mishra A, Jahoriya V, Veerasamy R, Tiwari AK, Rajak H. Targeting Lysosomes: A Strategy Against Chemoresistance in Cancer. Mini Rev Med Chem 2024; 24:1449-1468. [PMID: 38343053 DOI: 10.2174/0113895575287242240129120002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 01/11/2024] [Accepted: 01/19/2024] [Indexed: 07/23/2024]
Abstract
Chemotherapy is still the major method of treatment for many types of cancer. Curative cancer therapy is hampered significantly by medication resistance. Acidic organelles like lysosomes serve as protagonists in cellular digestion. Lysosomes, however, are gaining popularity due to their speeding involvement in cancer progression and resistance. For instance, weak chemotherapeutic drugs of basic nature permeate through the lysosomal membrane and are retained in lysosomes in their cationic state, while extracellular release of lysosomal enzymes induces cancer, cytosolic escape of lysosomal hydrolases causes apoptosis, and so on. Drug availability at the sites of action is decreased due to lysosomal drug sequestration, which also enhances cancer resistance. This review looks at lysosomal drug sequestration mechanisms and how they affect cancer treatment resistance. Using lysosomes as subcellular targets to combat drug resistance and reverse drug sequestration is another method for overcoming drug resistance that is covered in this article. The present review has identified lysosomal drug sequestration as one of the reasons behind chemoresistance. The article delves deeper into specific aspects of lysosomal sequestration, providing nuanced insights, critical evaluations, or novel interpretations of different approaches that target lysosomes to defect cancer.
Collapse
Affiliation(s)
- Ekta Shirbhate
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Vaibhav Singh
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Aditya Mishra
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Varsha Jahoriya
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| | - Ravichandran Veerasamy
- Faculty of Pharmacy, AIMST University, Semeling, 08100 Bedong, Kedah Darul Aman, Malaysia
| | - Amit K Tiwari
- UAMS College of Pharmacy; UAMS - University of Arkansas for Medical Sciences, (AR) USA
| | - Harish Rajak
- Department of Pharmacy, Guru Ghasidas University, Bilaspur-495 009, (C.G.), India
| |
Collapse
|
36
|
Deng Z, Chen S, Liu G, Zhu G. Unlocking the potential of platinum drugs: organelle-targeted small-molecule platinum complexes for improved anticancer performance. RSC Chem Biol 2023; 4:1003-1013. [PMID: 38033725 PMCID: PMC10685827 DOI: 10.1039/d3cb00087g] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 09/29/2023] [Indexed: 12/02/2023] Open
Abstract
Platinum-based drugs have revolutionized cancer chemotherapy; however, their therapeutic efficacy has been limited by severe side effects and drug resistance. Recently, approaches that target specific organelles in cancer cells have emerged as attractive alternatives to overcome these challenges. Many studies have validated these strategies and highlighted that organelle-targeted platinum complexes demonstrate increased anticancer activity, the ability to overcome drug resistance, novel molecular mechanisms, or even lower toxicity. This review provides a brief summary of various organelle-targeting strategies that promote the accumulation of platinum complexes in certain intracellular areas, such as the nucleus, mitochondria, endoplasmic reticulum (ER), and lysosomes. Moreover, the mechanisms through which these strategies improve anticancer performance, overcome drug resistance, and alter the action mode of conventional platinum drugs are discussed. By providing an extensive account of platinum complexes targeting different organelles, this review aims to assist researchers in understanding the design principles, identifying potential targets, and fostering innovative ideas for the development of platinum complexes.
Collapse
Affiliation(s)
- Zhiqin Deng
- Department of Chemistry, City University of Hong Kong Hong Kong SAR P. R. China
- City University of Hong Kong Shenzhen Research Institute Shenzhen 518057 P. R. China
- School of Medicine, Chongqing University Chongqing 400030 P. R. China
| | - Shu Chen
- Department of Chemistry, City University of Hong Kong Hong Kong SAR P. R. China
- City University of Hong Kong Shenzhen Research Institute Shenzhen 518057 P. R. China
| | - Gongyuan Liu
- Department of Chemistry, City University of Hong Kong Hong Kong SAR P. R. China
- City University of Hong Kong Shenzhen Research Institute Shenzhen 518057 P. R. China
| | - Guangyu Zhu
- Department of Chemistry, City University of Hong Kong Hong Kong SAR P. R. China
- City University of Hong Kong Shenzhen Research Institute Shenzhen 518057 P. R. China
| |
Collapse
|
37
|
Lin CP, Wu SH, Lin TY, Chu CH, Lo LW, Kuo CC, Chang JY, Hsu SC, Ko BS, Yao M, Hsiao JK, Wang SW, Huang DM. Lysosomal-targeted doxorubicin delivery using RBC-derived vesicles to overcome drug-resistant cancer through mitochondrial-dependent cell death. Pharmacol Res 2023; 197:106945. [PMID: 37797662 DOI: 10.1016/j.phrs.2023.106945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/30/2023] [Accepted: 10/02/2023] [Indexed: 10/07/2023]
Abstract
Multidrug resistance (MDR) is a major challenge in cancer chemotherapy. Nanoparticles as drug delivery systems (DDSs) show promise for MDR cancer therapy. However, current DDSs require sophisticated design and construction based on xenogeneic nanomaterials, evoking feasibility and biocompatibility concerns. Herein, a simple but versatile biological DDS (bDDS) composed of human red blood cell (RBC)-derived vesicles (RDVs) with excellent biocompatibility was surface-linked with doxorubicin (Dox) using glutaraldehyde (glu) to form Dox-gluRDVs that remarkably suppressed MDR in uterine sarcoma through a lysosomal-mitochondrial axis-dependent cell death mechanism. Dox-gluRDVs can efficiently deliver and accumulate Dox in lysosomes, bypassing drug efflux transporters and facilitating cellular uptake and retention of Dox in drug-resistant MES-SA/Dx5 cells. The transfer of lysosomal calcium to the mitochondria during mitochondria-lysosome contact due to lysosomal Dox accumulation may result in mitochondrial ROS overproduction, mitochondrial membrane potential loss, and activation of apoptotic signaling for the superior anti-MDR activity of Dox-gluRDVs in vitro and in vivo. This work highlights the great promise of RDVs to serve as a bDDS of Dox to overcome MDR cancers but also opens up a reliable strategy for lysosomal-mitochondrial axis-dependent cell death for fighting against other inoperable cancers.
Collapse
Affiliation(s)
- Chih-Peng Lin
- Department of Anesthesiology, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100225, Taiwan
| | - Shu-Hui Wu
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 30503, Taiwan
| | - Tzu-Yin Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 30503, Taiwan
| | - Chia-Hui Chu
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 30503, Taiwan
| | - Leu-Wei Lo
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 30503, Taiwan
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung 404333, Taiwan
| | - Jang-Yang Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli 35053, Taiwan; Taipei Cancer Center, Taipei Medical University Hospital, Taipei 110, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Szu-Chun Hsu
- Department of Laboratory Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100225, Taiwan
| | - Bor-Sheng Ko
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100225, Taiwan; Department of Hematological Oncology, National Taiwan University Cancer Center, Taipei 106037, Taiwan
| | - Ming Yao
- Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, National Taiwan University, Taipei 100225, Taiwan
| | - Jong-Kai Hsiao
- Department of Medical Imaging, Taipei Tzu Chi General Hospital, Buddhist Tzu-Chi Medical Foundation, New Taipei City 23142, Taiwan; School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Shih-Wei Wang
- Institute of Biomedical Sciences, Mackay Medical College, New Taipei City 252005, Taiwan; Department of Medicine, Mackay Medical College, New Taipei City 252005, Taiwan; School of Pharmacy, College of Pharmacy, Kaohsiung 807378, Taiwan
| | - Dong-Ming Huang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli 30503, Taiwan.
| |
Collapse
|
38
|
Jiang W, Tie Z, Yu C, Chen Y, Liu D, Li B. An engineered nanoplatform inhibiting energy metabolism and lysosomal activity of tumor cells to multiply cisplatin-based chemotherapy. Biomaterials 2023; 302:122354. [PMID: 37879187 DOI: 10.1016/j.biomaterials.2023.122354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 09/21/2023] [Accepted: 10/14/2023] [Indexed: 10/27/2023]
Abstract
Although inhibiting the energy metabolism of tumor cells has become an effective measure to enhance chemotherapy, tumor cells can still escape the lethal effect of chemotherapy by entering a dormancy state with low-energy expenditure. Herein, the glutathione (GSH)-responsive nanoplatform (C-A-D NPs) were constructed to inhibit energy metabolism and lysosomal activity of tumor cells, thereby forcing tumor cells to remain vulnerable to cisplatin. In this system, cisplatin prodrug was reduced to cisplatin by GSH, and D-peptide and apoptozole (Az) were released to inhibit the energy metabolism and autophagy-lysosome pathway of tumor cells. The suppressed autophagy-lysosome pathway prevents tumor cells from entering a low-energy dormancy state, resulting in the loss of resistance to the lethal effect of cisplatin with high-energy expenditure and insufficient energy supply. Such engineered nanoplatform effectively enhances the chemotherapeutic effect of cisplatin by inhibiting intracellular energy metabolism and lysosomal activity, showing great clinical prospects.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Biochemistry and Molecular Biology, Medical College, Guangxi University of Science and Technology, Liuzhou, 545005, China; College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing, 210033, China
| | - Zuoxiu Tie
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing, 210033, China
| | - Chi Yu
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, China
| | - Yu Chen
- Commonwealth Scientific and Industrial Research Organisation (CSIRO) Manufacturing, Clayton, Victoria, 3168, Australia
| | - Dan Liu
- College of Pharmaceutical and Biological Engineering, Shenyang University of Chemical Technology, Shenyang, 110142, China.
| | - Bin Li
- Department of Biochemistry and Molecular Biology, Medical College, Guangxi University of Science and Technology, Liuzhou, 545005, China.
| |
Collapse
|
39
|
Tejeda-Munoz N, Azbazdar Y, Monka J, Binder G, Dayrit A, Ayala R, O'Brien N, De Robertis EM. The PMA phorbol ester tumor promoter increases canonical Wnt signaling via macropinocytosis. eLife 2023; 12:RP89141. [PMID: 37902809 PMCID: PMC10615368 DOI: 10.7554/elife.89141] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023] Open
Abstract
Activation of the Wnt pathway lies at the core of many human cancers. Wnt and macropinocytosis are often active in the same processes, and understanding how Wnt signaling and membrane trafficking cooperate should improve our understanding of embryonic development and cancer. Here, we show that a macropinocytosis activator, the tumor promoter phorbol 12-myristate 13-acetate (PMA), enhances Wnt signaling. Experiments using the Xenopus embryo as an in vivo model showed marked cooperation between the PMA phorbol ester and Wnt signaling, which was blocked by inhibitors of macropinocytosis, Rac1 activity, and lysosome acidification. Human colorectal cancer tissue arrays and xenografts in mice showed a correlation of cancer progression with increased macropinocytosis/multivesicular body/lysosome markers and decreased GSK3 levels. The crosstalk between canonical Wnt, focal adhesions, lysosomes, and macropinocytosis suggests possible therapeutic targets for cancer progression in Wnt-driven cancers.
Collapse
Affiliation(s)
- Nydia Tejeda-Munoz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
- Department of Oncology Science, Health Stephenson Cancer Center, University of Oklahoma Health Science CenterOklahoma CityUnited States
| | - Yagmur Azbazdar
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Julia Monka
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Grace Binder
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Alex Dayrit
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Raul Ayala
- Department of Medicine, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Neil O'Brien
- Department of Medicine, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| | - Edward M De Robertis
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los AngelesLos AngelesUnited States
| |
Collapse
|
40
|
Verhagen NE, Koenderink JB, Blijlevens NMA, Janssen JJWM, Russel FGM. Transporter-Mediated Cellular Distribution of Tyrosine Kinase Inhibitors as a Potential Resistance Mechanism in Chronic Myeloid Leukemia. Pharmaceutics 2023; 15:2535. [PMID: 38004514 PMCID: PMC10675650 DOI: 10.3390/pharmaceutics15112535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023] Open
Abstract
Chronic myeloid leukemia (CML) is a hematologic neoplasm characterized by the expression of the BCR::ABL1 oncoprotein, a constitutively active tyrosine kinase, resulting in uncontrolled growth and proliferation of cells in the myeloid lineage. Targeted therapy using tyrosine kinase inhibitors (TKIs) such as imatinib, nilotinib, dasatinib, bosutinib, ponatinib and asciminib has drastically improved the life expectancy of CML patients. However, treatment resistance occurs in 10-20% of CML patients, which is a multifactorial problem that is only partially clarified by the presence of TKI inactivating BCR::ABL1 mutations. It may also be a consequence of a reduction in cytosolic TKI concentrations in the target cells due to transporter-mediated cellular distribution. This review focuses on drug-transporting proteins in stem cells and progenitor cells involved in the distribution of TKIs approved for the treatment of CML. Special attention will be given to ATP-binding cassette transporters expressed in lysosomes, which may facilitate the extracytosolic sequestration of these compounds.
Collapse
Affiliation(s)
- Noor E. Verhagen
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.E.V.); (J.B.K.)
| | - Jan B. Koenderink
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.E.V.); (J.B.K.)
| | - Nicole M. A. Blijlevens
- Department of Haematology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.M.A.B.); (J.J.W.M.J.)
| | - Jeroen J. W. M. Janssen
- Department of Haematology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.M.A.B.); (J.J.W.M.J.)
| | - Frans G. M. Russel
- Division of Pharmacology and Toxicology, Department of Pharmacy, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (N.E.V.); (J.B.K.)
| |
Collapse
|
41
|
Wen Q, Xie X, Chen C, Wen B, Liu Y, Zhou J, Lin X, Jin H, Shi K. Lipid reprogramming induced by the NNMT-ABCA1 axis enhanced membrane fluidity to promote endometrial cancer progression. Aging (Albany NY) 2023; 15:11860-11874. [PMID: 37889548 PMCID: PMC10683614 DOI: 10.18632/aging.205142] [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: 04/25/2023] [Accepted: 09/08/2023] [Indexed: 10/28/2023]
Abstract
Elucidating the mechanism for the high metastasis capacity of Endometrial cancer (EC) is crucial to improve treatment outcomes of EC. We have recently reported that nicotinamide N-methyltransferase (NNMT) is overexpressed in EC, especially in EC, and predicts poor survival of chemotherapy patients. Here, we aimed to determine the function and mechanism of NNMT on metastasis of EC. Additionally, analysis of public datasets indicated that NNMT is involved in cholesterol metabolism. In vitro, NNMT overexpression promoted migration and invasion of EC by reducing cholesterol levels in the cytoplasm and cell membrane. Mechanistically, NNMT activated ABCA1 expression, leading to cholesterol efflux and membrane fluidity enhancement, thereby promoting EC's epithelial-mesenchymal transition (EMT). In vivo, the metastasis capacity of EC was weakened by targeting NNMT. Our findings suggest a new molecular mechanism involving NNMT in metastasis, poor survival of EC mediated by PP2A and affecting cholesterol metabolism.
Collapse
Affiliation(s)
- Qirong Wen
- Department of Gynecology and Obstetrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaohui Xie
- Department of Gynecology and Obstetrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Caiyuan Chen
- Prenatal Diagnosis Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
- Department of Obstetrics and Gynecology, Guangzhou Medical University, Guangzhou, China
| | - Bolun Wen
- Department of Gynecology and Obstetrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yaqiong Liu
- Department of Gynecology and Obstetrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jie Zhou
- Department of Gynecology and Obstetrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaobin Lin
- Department of Breast Surgery and General Surgery, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Han Jin
- Prenatal Diagnosis Center, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Kun Shi
- Department of Gynecology and Obstetrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
42
|
Yang J, Li L, Cheng J, Lu J, Zhang S, Wang S, Zhao L, Zhou L. The m6A modulator-mediated cytarabine sensitivity and immune cell infiltration signature in acute myeloid leukemia. J Cancer Res Clin Oncol 2023; 149:11457-11469. [PMID: 37391640 DOI: 10.1007/s00432-023-05029-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/26/2023] [Indexed: 07/02/2023]
Abstract
PURPOSE The study aims to investigate the impact of m6A modulators on drug resistance and the immune microenvironment in acute myeloid leukemia (AML). The emergence of drug resistance is a significant factor that contributes to relapse and refractory AML, leading to a poor prognosis. METHODS The AML transcriptome data were retrieved from the TCGA database. The "oncoPredict" R package was utilized to assess the sensitivity of each sample to cytarabine (Ara-C) and classify them into distinct groups. Differential expression analysis was performed to identify m6A modulators differentially expressed between the two groups. Select Random Forest (RF) to build a predictive model. Model performance was evaluated using calibration curve, clinical decision curve, and clinical impact curve. The impacts of METTL3 on Ara-C sensitivity and immune microenvironment in AML were examined using GO, KEGG, CIBERSORT, and GSEA analyses. RESULTS Seventeen out of 26 m6A modulators exhibited differential expression between the Ara-C-sensitive and resistant groups, with a high degree of correlation. We selected the 5 genes with the highest scores in the RF model to build a reliable and accurate prediction model. METTL3 plays a vital role in m6A modification, and further analysis shows its impact on the sensitivity of AML cells to Ara-C through its interaction with 7 types of immune-infiltrating cells and autophagy. CONCLUSION This study utilizes m6A modulators to develop a prediction model for the sensitivity of AML patients to Ara-C, which can assist in treating AML drug resistance by targeting mRNA methylation.
Collapse
Affiliation(s)
- Jincai Yang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Liangliang Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, 730000, Gansu, China
| | - Juan Cheng
- Department of Hematology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Jianle Lu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Shuling Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Shan Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Li Zhao
- Central Laboratory, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China.
- Gansu Key Laboratory of Genetic Study of Hematopathy, Lanzhou, 730000, Gansu, China.
| | - Lanxia Zhou
- Central Laboratory, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu, China.
- Gansu Key Laboratory of Genetic Study of Hematopathy, Lanzhou, 730000, Gansu, China.
| |
Collapse
|
43
|
Han H, Xing L, Chen BT, Liu Y, Zhou TJ, Wang Y, Zhang LF, Li L, Cho CS, Jiang HL. Progress on the pathological tissue microenvironment barrier-modulated nanomedicine. Adv Drug Deliv Rev 2023; 200:115051. [PMID: 37549848 DOI: 10.1016/j.addr.2023.115051] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 07/21/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Imbalance in the tissue microenvironment is the main obstacle to drug delivery and distribution in the human body. Before penetrating the pathological tissue microenvironment to the target site, therapeutic agents are usually accompanied by three consumption steps: the first step is tissue physical barriers for prevention of their penetration, the second step is inactivation of them by biological molecules, and the third step is a cytoprotective mechanism for preventing them from functioning on specific subcellular organelles. However, recent studies in drug-hindering mainly focus on normal physiological rather than pathological microenvironment, and the repair of damaged physiological barriers is also rarely discussed. Actually, both the modulation of pathological barriers and the repair of damaged physiological barriers are essential in the disease treatment and the homeostasis maintenance. In this review, we present an overview describing the latest advances in the generality of these pathological barriers and barrier-modulated nanomedicine. Overall, this review holds considerable significance for guiding the design of nanomedicine to increase drug efficacy in the future.
Collapse
Affiliation(s)
- Han Han
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Lei Xing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China
| | - Bi-Te Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Tian-Jiao Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Yi Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling-Feng Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China
| | - Ling Li
- Department of Endocrinology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Korea.
| | - Hu-Lin Jiang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China; College of Pharmacy, Yanbian University, Yanji 133002, China.
| |
Collapse
|
44
|
He X, Li X, Tian W, Li C, Li P, Zhao J, Yang S, Li S. The role of redox-mediated lysosomal dysfunction and therapeutic strategies. Biomed Pharmacother 2023; 165:115121. [PMID: 37418979 DOI: 10.1016/j.biopha.2023.115121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/30/2023] [Accepted: 07/01/2023] [Indexed: 07/09/2023] Open
Abstract
Redox homeostasis refers to the dynamic equilibrium between oxidant and reducing agent in the body which plays a crucial role in maintaining normal physiological activities of the body. The imbalance of redox homeostasis can lead to the development of various human diseases. Lysosomes regulate the degradation of cellular proteins and play an important role in influencing cell function and fate, and lysosomal dysfunction is closely associated with the development of various diseases. In addition, several studies have shown that redox homeostasis plays a direct or indirect role in regulating lysosomes. Therefore, this paper systematically reviews the role and mechanisms of redox homeostasis in the regulation of lysosomal function. Therapeutic strategies based on the regulation of redox exerted to disrupt or restore lysosomal function are further discussed. Uncovering the role of redox in the regulation of lysosomes helps to point new directions for the treatment of many human diseases.
Collapse
Affiliation(s)
- Xiaomeng He
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xuening Li
- Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Wei Tian
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Chenyu Li
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Pengfei Li
- The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jingyuan Zhao
- The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Shilei Yang
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Shuai Li
- Department of Pharmacy, The First Affiliated Hospital of Dalian Medical University, Dalian, China.
| |
Collapse
|
45
|
Tejeda-Muñoz N, Azbazdar Y, Monka J, Binder G, Dayrit A, Ayala R, O’Brien N, De Robertis EM. The PMA Phorbol Ester Tumor Promoter Increases Canonical Wnt Signaling Via Macropinocytosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.02.543509. [PMID: 37333286 PMCID: PMC10274750 DOI: 10.1101/2023.06.02.543509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Activation of the Wnt pathway lies at the core of many human cancers. Wnt and macropinocytosis are often active in the same processes, and understanding how Wnt signaling and membrane trafficking cooperate should improve our understanding of embryonic development and cancer. Here we show that a macropinocytosis activator, the tumor promoter Phorbol 12-myristate 13-acetate (PMA), enhances Wnt signaling. Experiments using the Xenopus embryo as an in vivo model showed marked cooperation between the PMA phorbol ester and Wnt signaling, which was blocked by inhibitors of macropinocytosis, Rac1 activity, and lysosome acidification. Human colorectal cancer tissue arrays and xenografts in mice showed a correlation of cancer progression with increased macropinocytosis/multivesicular body/lysosome markers and decreased GSK3 levels. The crosstalk between canonical Wnt, focal adhesions, lysosomes, and macropinocytosis suggests possible therapeutic targets for cancer progression in Wnt-driven cancers.
Collapse
Affiliation(s)
- Nydia Tejeda-Muñoz
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
- Department of Oncology Science, Health Stephenson Cancer Center, University of Oklahoma Health Science Center, Oklahoma City, Oklahoma 73104, USA
- These authors contributed equally
| | - Yagmur Azbazdar
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
- These authors contributed equally
| | - Julia Monka
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
| | - Grace Binder
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
| | - Alex Dayrit
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
| | - Raul Ayala
- Department of Medicine, David Geffen School of Medicine at UCLA
| | - Neil O’Brien
- Department of Medicine, David Geffen School of Medicine at UCLA
| | - Edward M. De Robertis
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, 90095-1662, USA
| |
Collapse
|
46
|
Gao HL, Cui Q, Wang JQ, Ashby CR, Chen Y, Shen ZX, Chen ZS. The AKT inhibitor, MK-2206, attenuates ABCG2-mediated drug resistance in lung and colon cancer cells. Front Pharmacol 2023; 14:1235285. [PMID: 37521473 PMCID: PMC10373739 DOI: 10.3389/fphar.2023.1235285] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/03/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction: The overexpression of ATP-binding cassette (ABC) transporters, ABCB1 and ABCG2, are two of the major mediators of multidrug resistance (MDR) in cancers. Although multiple ABCB1 and ABCG2 inhibitors have been developed and some have undergone evaluation in clinical trials, none have been clinically approved. The compound, MK-2206, an inhibitor of the protein kinases AKT1/2/3, is undergoing evaluation in multiple clinical trials for the treatment of certain types of cancers, including those resistant to erlotinib. In this in vitro study, we conducted in vitro experiments to determine if MK-2206 attenuates multidrug resistance in cancer cells overexpressing the ABCB1 or ABCG2 transporter. Methodology: The efficacy of MK-2206 (0.03-1 μM), in combination with the ABCB1 transporter sub-strates doxorubicin and paclitaxel, and ABCG2 transporter substrates mitoxantrone, SN-38 and topotecan, were determined in the cancer cell lines, KB-C2 and SW620/Ad300, which overexpress the ABCB1 transporter or H460/MX20 and S1-M1-80, which overexpress the ABCG2 transporter, respectively. The expression level and the localization of ABCG2 transporter on the cancer cells membranes were determined using western blot and immunofluorescence assays, respectively, following the incubation of cells with MK-2206. Finally, the interaction between MK-2206 and human ABCG2 transporter was predicted using computer-aided molecular modeling. Results: MK-2206 significantly increased the efficacy of anticancer compounds that were substrates for the ABCG2 but not the ABCB1 transporter. MK-2206 alone (0.03-1 μM) did not significantly alter the viability of H460/MX20 and S1-M1-80 cancer cells, which overexpress the ABCG2 transporter, compared to cells incubated with vehicle. However, MK-2206 (0.3 and 1 μM) significantly increased the anticancer efficacy of mitoxantrone, SN-38 and topotecan, in H460/MX20 and S1-M1-80 cancer cells, as indicated by a significant decrease in their IC50 values, compared to cells incubated with vehicle. MK-2206 significantly increased the basal activity of the ABCG2 ATPase (EC50 = 0.46 μM) but did not significantly alter its expression level and sub-localization in the membrane. The molecular modeling results suggested that MK-2206 binds to the active pocket of the ABCG2 transporter, by a hydrogen bond, hydrophobic interactions and π-π stacking. Conclusion: These in vitro data indicated that MK-2206 surmounts resistance to mitoxantrone, SN-38 and topotecan in cancer cells overexpressing the ABCG2 transporter. If these results can be translated to humans, it is possible that MK-2206 could be used to surmount MDR in cancer cells overexpressing the ABCG2 transporter.
Collapse
Affiliation(s)
- Hai-Ling Gao
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Qingbin Cui
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Charles R. Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Yanchun Chen
- Department of Histology and Embryology, Weifang Medical University, Weifang, Shandong, China
| | - Zhi-Xin Shen
- Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| |
Collapse
|
47
|
Wang Y, Chen YY, Gao GB, Zheng YH, Yu NN, Ouyang L, Gao X, Li N, Wen SY, Huang S, Zhao Q, Liu L, Cao M, Zhang S, Zhang J, He QY. Polyphyllin D punctures hypertrophic lysosomes to reverse drug resistance of hepatocellular carcinoma by targeting acid sphingomyelinase. Mol Ther 2023; 31:2169-2187. [PMID: 37211762 PMCID: PMC10362416 DOI: 10.1016/j.ymthe.2023.05.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 04/13/2023] [Accepted: 05/18/2023] [Indexed: 05/23/2023] Open
Abstract
Hypertrophic lysosomes are critical for tumor progression and drug resistance; however, effective and specific lysosome-targeting compounds for cancer therapy are lacking. Here we conducted a lysosomotropic pharmacophore-based in silico screen in a natural product library (2,212 compounds), and identified polyphyllin D (PD) as a novel lysosome-targeted compound. PD treatment was found to cause lysosomal damage, as evidenced by the blockade of autophagic flux, loss of lysophagy, and the release of lysosomal contents, thus exhibiting anticancer effects on hepatocellular carcinoma (HCC) cell both in vitro and in vivo. Closer mechanistic examination revealed that PD suppressed the activity of acid sphingomyelinase (SMPD1), a lysosomal phosphodieserase that catalyzes the hydrolysis of sphingomyelin to produce ceramide and phosphocholine, by directly occupying its surface groove, with Trp148 in SMPD1 acting as a major binding residue; this suppression of SMPD1 activity irreversibly triggers lysosomal injury and initiates lysosome-dependent cell death. Furthermore, PD-enhanced lysosomal membrane permeabilization to release sorafenib, augmenting the anticancer effect of sorafenib both in vivo and in vitro. Overall, our study suggests that PD can potentially be further developed as a novel autophagy inhibitor, and a combination of PD with classical chemotherapeutic anticancer drugs could represent a novel therapeutic strategy for HCC intervention.
Collapse
Affiliation(s)
- Yang Wang
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China.
| | - Yan-Yan Chen
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Gui-Bin Gao
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yang-Han Zheng
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Nan-Nan Yu
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Lan Ouyang
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xuejuan Gao
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Nan Li
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Shi-Yuan Wen
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Shangjia Huang
- MOE Key Laboratory of Tumor Molecular Biology, The First Affiliated Hospital of Jinan University, Guangzhou 510613, China
| | - Qian Zhao
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Langxia Liu
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Mingrong Cao
- Department of General Surgery, The First Affiliated Hospital, Jinan University, Guangzhou 510613, China
| | - Shuixing Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510613, China; MOE Key Laboratory of Tumor Molecular Biology, The First Affiliated Hospital of Jinan University, Guangzhou 510613, China.
| | - Jing Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou 510613, China; MOE Key Laboratory of Tumor Molecular Biology, The First Affiliated Hospital of Jinan University, Guangzhou 510613, China.
| | - Qing-Yu He
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou 510632, China; MOE Key Laboratory of Tumor Molecular Biology, The First Affiliated Hospital of Jinan University, Guangzhou 510613, China.
| |
Collapse
|
48
|
Ferret L, Alvarez-Valadez K, Rivière J, Muller A, Bohálová N, Yu L, Guittat L, Brázda V, Kroemer G, Mergny JL, Djavaheri-Mergny M. G-quadruplex ligands as potent regulators of lysosomes. Autophagy 2023; 19:1901-1915. [PMID: 36740766 PMCID: PMC10283436 DOI: 10.1080/15548627.2023.2170071] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/03/2022] [Accepted: 12/05/2022] [Indexed: 02/07/2023] Open
Abstract
Guanine-quadruplex structures (G4) are unusual nucleic acid conformations formed by guanine-rich DNA and RNA sequences and known to control gene expression mechanisms, from transcription to protein synthesis. So far, a number of molecules that recognize G4 have been developed for potential therapeutic applications in human pathologies, including cancer and infectious diseases. These molecules are called G4 ligands. When the biological effects of G4 ligands are studied, the analysis is often limited to nucleic acid targets. However, recent evidence indicates that G4 ligands may target other cellular components and compartments such as lysosomes and mitochondria. Here, we summarize our current knowledge of the regulation of lysosome by G4 ligands, underlying their potential functional impact on lysosome biology and autophagic flux, as well as on the transcriptional regulation of lysosomal genes. We outline the consequences of these effects on cell fate decisions and we systematically analyzed G4-prone sequences within the promoter of 435 lysosome-related genes. Finally, we propose some hypotheses about the mechanisms involved in the regulation of lysosomes by G4 ligands.
Collapse
Affiliation(s)
- Lucille Ferret
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Equipe labellisée par la Ligue contre le Cancer, Institut universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Karla Alvarez-Valadez
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Equipe labellisée par la Ligue contre le Cancer, Institut universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Jennifer Rivière
- Department of Medicine III, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Alexandra Muller
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Equipe labellisée par la Ligue contre le Cancer, Institut universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| | - Natalia Bohálová
- Department of Biophysical Chemistry and Molecular Oncology, Institute of Biophysics, The Czech Academy of Sciences, Brno, Czech Republic
| | - Luo Yu
- Laboratoire d’Optique et Biosciences, Ecole Polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, 91128Palaiseau, France
- CNRS UMR9187, INSERM U1196, Université Paris-Saclay, Orsay, France
| | - Lionel Guittat
- Laboratoire d’Optique et Biosciences, Ecole Polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, 91128Palaiseau, France
- UFR SMBH, Université Sorbonne Paris Nord, Bobigny, France
| | - Vaclav Brázda
- Department of Biophysical Chemistry and Molecular Oncology, Institute of Biophysics, The Czech Academy of Sciences, Brno, Czech Republic
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Equipe labellisée par la Ligue contre le Cancer, Institut universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Jean-Louis Mergny
- Department of Biophysical Chemistry and Molecular Oncology, Institute of Biophysics, The Czech Academy of Sciences, Brno, Czech Republic
- Laboratoire d’Optique et Biosciences, Ecole Polytechnique, CNRS, INSERM, Institut Polytechnique de Paris, 91128Palaiseau, France
| | - Mojgan Djavaheri-Mergny
- Centre de Recherche des Cordeliers, INSERM UMRS 1138, Sorbonne Université, Université Paris Cité, Equipe labellisée par la Ligue contre le Cancer, Institut universitaire de France, Paris, France
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
| |
Collapse
|
49
|
Yuan Y, Olawode EO, Tumey LN, Lu F. Visualizing drug-induced lipid accumulation in lysosomes of live cancer cells with stimulated Raman imaging. BIOMEDICAL OPTICS EXPRESS 2023; 14:2551-2564. [PMID: 37342714 PMCID: PMC10278636 DOI: 10.1364/boe.487527] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 06/23/2023]
Abstract
The low pH of the lysosomal compartment often results in sequestration of chemotherapeutic agents that contain positively charged basic functional groups, leading to anti-cancer drug resistance. To visualize drug localization in lysosomes and its influence on lysosomal functions, we synthesize a group of drug-like compounds that contain both a basic functional group and a bisarylbutadiyne (BADY) group as a Raman probe. With quantitative stimulated Raman scattering (SRS) imaging, we validate that the synthesized lysosomotropic (LT) drug analogs show high lysosomal affinity, which can also serve as a photostable lysosome tracker. We find that long-term retention of the LT compounds in lysosomes leads to the increased amount and colocalization of both lipid droplets (LDs) and lysosomes in SKOV3 cells. With hyperspectral SRS imaging, further studies find that the LDs stuck in lysosomes are more saturated than the LDs staying out of the lysosomes, indicating impaired lysosomal lipid metabolism by the LT compounds. These results demonstrate that SRS imaging of the alkyne-based probes is a promising approach to characterizing the lysosomal sequestration of drugs and its influence on cell functions.
Collapse
Affiliation(s)
- Yuhao Yuan
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York, Binghamton, NY 13902, USA
- Current Address: Department of Electrical and Computer Engineering, Boston University, Boston, MA 02215, USA
| | - Emmanuel O. Olawode
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, State University of New York, Binghamton, NY 13902, USA
- Current Address: College of Pharmacy, Larkin University, Miami, FL 33169, USA
| | - L. Nathan Tumey
- School of Pharmacy and Pharmaceutical Sciences, Binghamton University, State University of New York, Binghamton, NY 13902, USA
| | - Fake Lu
- Department of Biomedical Engineering, Thomas J. Watson College of Engineering and Applied Science, Binghamton University, State University of New York, Binghamton, NY 13902, USA
| |
Collapse
|
50
|
Lei Z, Tian Q, Teng Q, Wurpel JND, Zeng L, Pan Y, Chen Z. Understanding and targeting resistance mechanisms in cancer. MedComm (Beijing) 2023; 4:e265. [PMID: 37229486 PMCID: PMC10203373 DOI: 10.1002/mco2.265] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 03/05/2023] [Accepted: 03/23/2023] [Indexed: 05/27/2023] Open
Abstract
Resistance to cancer therapies has been a commonly observed phenomenon in clinical practice, which is one of the major causes of treatment failure and poor patient survival. The reduced responsiveness of cancer cells is a multifaceted phenomenon that can arise from genetic, epigenetic, and microenvironmental factors. Various mechanisms have been discovered and extensively studied, including drug inactivation, reduced intracellular drug accumulation by reduced uptake or increased efflux, drug target alteration, activation of compensatory pathways for cell survival, regulation of DNA repair and cell death, tumor plasticity, and the regulation from tumor microenvironments (TMEs). To overcome cancer resistance, a variety of strategies have been proposed, which are designed to enhance the effectiveness of cancer treatment or reduce drug resistance. These include identifying biomarkers that can predict drug response and resistance, identifying new targets, developing new targeted drugs, combination therapies targeting multiple signaling pathways, and modulating the TME. The present article focuses on the different mechanisms of drug resistance in cancer and the corresponding tackling approaches with recent updates. Perspectives on polytherapy targeting multiple resistance mechanisms, novel nanoparticle delivery systems, and advanced drug design tools for overcoming resistance are also reviewed.
Collapse
Affiliation(s)
- Zi‐Ning Lei
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Qin Tian
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
| | - Qiu‐Xu Teng
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - John N. D. Wurpel
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Leli Zeng
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
| | - Yihang Pan
- PrecisionMedicine CenterScientific Research CenterThe Seventh Affiliated HospitalSun Yat‐Sen UniversityShenzhenP. R. China
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNew YorkUSA
| |
Collapse
|