1
|
Vangala V, Chen YC, Dinavahi SS, Gowda K, Lone NA, Herlyn M, Drabick J, Helm K, Zhu J, Neves RI, Sharma AK, Berg A, Archetti M, Amin S, Schell TD, Robertson GP. Tumor Heterogeneity Shapes Survival Dynamics in Drug-Treated Cells, Revealing Size-Drifting Subpopulations. ACS Pharmacol Transl Sci 2024; 7:3573-3584. [PMID: 39539277 PMCID: PMC11555517 DOI: 10.1021/acsptsci.4c00453] [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: 07/25/2024] [Revised: 09/26/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
The goal of this project was to demonstrate that subpopulations of cells in tumors can uniquely fluctuate in size in response to environmental conditions created during drug treatment, thereby acting as a dynamic "rheostat" to create a favorable tumor environment for growth. The cancer modeling used for these studies was subpopulations of melanoma cells existing in cultured and tumor systems that differed in aldehyde dehydrogenase (ALDH) activity. However, similar observations were found in other cancer types in addition to melanoma, making them applicable broadly across cancer. The approach was designed to show that either ALDHhigh and ALDHlow subpopulations rapidly epigenetically transition between stem-cell-like high into nonstem-like low production states to create an environment during drug treatment that would enable optimal cellular proliferation and tumor expansion to facilitate drug resistance. The controlled experiments showed proportional changes in each cell population to reach an evolutionarily stable equilibrium mediated by the needed levels of ALDH enzyme activity. Mechanistically, cell population size changes served to functionally move the aldehyde and the resulting reactive oxygen species (ROS) levels to those compatible with optimal cellular proliferation with population fluctuations dependent on the levels of drug induced tumor stress. This is the first report documenting fluctuations in the sizes of cell populations in tumors to cooperatively assist in drug resistance development.
Collapse
Affiliation(s)
- Venugopal Vangala
- Department
of Pharmacology, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Yu-Chi Chen
- Department
of Pharmacology, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Saketh S. Dinavahi
- Department
of Pharmacology, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Krishne Gowda
- Department
of Pharmacology, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Nazir A. Lone
- Department
of Pharmacology, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Meenhard Herlyn
- Molecular
and Cellular Oncogenesis Program and Melanoma Research Center, Wistar Institute, Philadelphia, Pennsylvania 19104, United States
| | - Joseph Drabick
- Department
of Medicine, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Klaus Helm
- Department
of Dermatology, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Jiyue Zhu
- Department
of Pharmaceutical Sciences, Washington State
University College of Pharmacy and Pharmaceutical Sciences, Spokane, Washington 99202, United States
| | - Rogerio I. Neves
- Department
of Surgery, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Arun K. Sharma
- Department
of Pharmacology, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Arthur Berg
- Department
of Public Health Sciences, Pennsylvania
State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Marco Archetti
- Department
of Biology, Pennsylvania State University, State College, Pennsylvania 16802, United States
| | - Shantu Amin
- Department
of Pharmacology, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Todd D. Schell
- Department
of Microbiology and Immunology, Pennsylvania
State University College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Gavin P. Robertson
- Department
of Pharmacology, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
- Department
of Dermatology, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
- Department
of Pathology, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
- Department
of Surgery, Pennsylvania State University
College of Medicine, Hershey, Pennsylvania 17033, United States
- Foreman
Foundation for Melanoma Research, Pennsylvania
State University College of Medicine, Hershey, Pennsylvania 17033, United States
- Melanoma
Center, Pennsylvania State University College
of Medicine, Hershey, Pennsylvania 17033, United States
- Melanoma
Therapeutics Program, Pennsylvania State
University College of Medicine, Hershey, Pennsylvania 17033, United States
| |
Collapse
|
2
|
Manouchehri JM, Datta J, Marcho LM, Reardon JJ, Stover D, Wesolowski R, Borate U, Cheng TYD, Schnell PM, Ramaswamy B, Sizemore GM, Rubinstein MP, Cherian MA. The role of heparan sulfate in enhancing the chemotherapeutic response in triple-negative breast cancer. Breast Cancer Res 2024; 26:153. [PMID: 39506780 PMCID: PMC11539583 DOI: 10.1186/s13058-024-01906-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/17/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND Breast cancer, one of the most common forms of cancer, is associated with the highest cancer-related mortality among women worldwide. In comparison to other types of breast cancer, patients diagnosed with the triple-negative breast cancer (TNBC) subtype have the worst outcome because current therapies do not produce long-lasting responses. Hence, innovative therapies that produce persisting responses are a critical need. We previously discovered that hyperactivating purinergic receptors (P2RXs) by increasing extracellular adenosine triphosphate (eATP) concentrations enhances TNBC cell lines' response to chemotherapy. Heparan sulfate inhibits multiple extracellular ATPases, so it is a molecule of interest in this regard. In turn, heparanase degrades polysulfated polysaccharide heparan sulfate. Importantly, previous work suggests that breast cancer and other cancers express heparanase at high levels. Hence, as heparan sulfate can inhibit extracellular ATPases to facilitate eATP accumulation, it may intensify responses to chemotherapy. We postulated that heparanase inhibitors would exacerbate chemotherapy-induced decreases in TNBC cell viability by increasing heparan sulfate in the cellular microenvironment and hence, augmenting eATP. METHODS We treated TNBC cell lines MDA-MB 231, Hs 578t, and MDA-MB 468 and non-tumorigenic immortal mammary epithelial MCF-10A cells with paclitaxel (cytotoxic chemotherapeutic) with or without the heparanase inhibitor OGT 2115 and/or supplemental heparan sulfate. We evaluated cell viability and the release of eATP. Also, we compared the expression of heparanase protein in cell lines and tissues by immunoblot and immunohistochemistry, respectively. In addition, we examined breast-cancer-initiating cell populations using tumorsphere formation efficiency assays on treated cells. RESULTS We found that combining heparanase inhibitor OGT 2115 with chemotherapy decreased TNBC cell viability and tumorsphere formation through increases in eATP and activation of purinergic receptors as compared to TNBC cells treated with single-agent paclitaxel. CONCLUSION Our data shows that by preventing heparan sulfate breakdown, heparanase inhibitors make TNBC cells more susceptible to chemotherapy by enhancing eATP concentrations.
Collapse
Affiliation(s)
- Jasmine M Manouchehri
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Jharna Datta
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Lynn M Marcho
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Jesse J Reardon
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Daniel Stover
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Robert Wesolowski
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Uma Borate
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Ting-Yuan David Cheng
- Division of Cancer Prevention and Control, Department of Internal Medicine, The Ohio State University, Suite 525, 1590 North High St., Columbus, OH, 43201, USA
| | - Patrick M Schnell
- Division of Biostatistics, The Ohio State University College of Public Health, 1841 Neil Ave., Columbus, OH, 43210, USA
| | - Bhuvaneswari Ramaswamy
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Gina M Sizemore
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Mark P Rubinstein
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA
| | - Mathew A Cherian
- Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, 410 W 10th Ave., Columbus, OH, 43210, USA.
- Division of Medical Oncology, 460 W 12th Ave., 888 BRT, Columbus, OH, 43210, USA.
| |
Collapse
|
3
|
Barranco MM, Zecchinati F, Perdomo VG, Habib MJ, Rico MJ, Rozados VR, Salazar M, Fusini ME, Scharovsky OG, Villanueva SSM, Mainetti LE, García F. Intestinal ABC transporters: Influence on the metronomic cyclophosphamide-induced toxic effect in an obese mouse mammary cancer model. Toxicol Appl Pharmacol 2024; 492:117130. [PMID: 39426530 DOI: 10.1016/j.taap.2024.117130] [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: 01/18/2024] [Revised: 09/26/2024] [Accepted: 10/16/2024] [Indexed: 10/21/2024]
Abstract
Metronomic chemotherapy (MCT) is a cancer therapeutic approach characterized by low dose drug chronic administration and limited or null toxicity. Obesity-induced metabolic alterations worsen cancer prognosis and influence the intestinal biochemical barrier, altering the Multidrug resistance-associated protein 2 (Mrp2) and Multidrug resistance protein-1 (Mdr-1), efflux pumps that transport chemotherapeutic drugs. Obesity and cancer are frequent co-morbidities; thus, our aim was to evaluate the effectiveness and toxicity of MCT with cyclophosphamide (Cy) in obese mice with metabolic alterations bearing a mammary adenocarcinoma. Simultaneously, the expression and activities of intestinal Mrp2 and Mdr-1 were assessed. CBi male mice, were fed with chow diet (C) or diet with 40 % of fat (HFD). After 16 weeks, metabolic alterations were confirmed by biochemical and morphological parameters. At that time-point, HFD group showed decreased expressions of Mrp2 mRNA (53 %) as well as Mdr-1a and Mdr-1b (42 % and 59 %, respectively), compared to C (P < 0.05). This result correlated with decreased intestinal Mrp2 and Mdr-1 efflux activities (64 % and 45 %, respectively), compared to C (P < 0.05). Ultimately, mice were challenged with M-406 mammary adenocarcinoma; when the tumor was palpable, mice were distributed into 4 groups. The % inhibition of tumor growth with Cy (30 mg/kg/day) in C + Cy was higher than that of HFD + Cy (P = 0.052). Besides, it was observed a 21 % diminution in body weight and leukopenia in the HFD + Cy group. Conclusion: Obesity-induced metabolic alterations impair intestinal Mrp2 and Mdr-1 functions, bringing about increments in Cy absorption, leading to toxicity; in addition, the antitumor effectiveness of MCT decreased in obese animals.
Collapse
MESH Headings
- Animals
- Cyclophosphamide/toxicity
- Mice
- Obesity/metabolism
- Male
- Female
- Administration, Metronomic
- Multidrug Resistance-Associated Protein 2
- Antineoplastic Agents, Alkylating/toxicity
- Mammary Neoplasms, Experimental/metabolism
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/chemically induced
- Mice, Obese
- Multidrug Resistance-Associated Proteins/metabolism
- Multidrug Resistance-Associated Proteins/genetics
- Intestinal Mucosa/drug effects
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Adenocarcinoma/pathology
- Adenocarcinoma/drug therapy
- Adenocarcinoma/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP-Binding Cassette Transporters/metabolism
- ATP-Binding Cassette Transporters/genetics
- Diet, High-Fat
Collapse
Affiliation(s)
- María Manuela Barranco
- Laboratorio de Fisiología Metabólica, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina.; CONICET-Rosario. Rosario, Santa Fe, Argentina
| | - Felipe Zecchinati
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Fisiología Experimental-CONICET. Rosario, Santa Fe, Argentina
| | - Virginia Gabriela Perdomo
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Facultad de Ciencias Bioquímicas y Farmacéuticas-Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Martín José Habib
- Laboratorio de Fisiología Metabólica, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - María José Rico
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Viviana Rosa Rozados
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Mario Salazar
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Laboratorio de Farmacognosia, Facultad de Ciencias Bioquímicas y Farmacéuticas Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Matías Ezequiel Fusini
- Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Olga Graciela Scharovsky
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina.; CIC-UNR, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | | | - Leandro Ernesto Mainetti
- CONICET-Rosario. Rosario, Santa Fe, Argentina.; Instituto de Genética Experimental, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina
| | - Fabiana García
- Laboratorio de Fisiología Metabólica, Facultad de Ciencias Médicas, Universidad Nacional de Rosario. Rosario, Santa Fe, Argentina.; CONICET-Rosario. Rosario, Santa Fe, Argentina..
| |
Collapse
|
4
|
Lavudi K, Nuguri SM, Pandey P, Kokkanti RR, Wang QE. ALDH and cancer stem cells: Pathways, challenges, and future directions in targeted therapy. Life Sci 2024; 356:123033. [PMID: 39222837 DOI: 10.1016/j.lfs.2024.123033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/16/2024] [Accepted: 08/30/2024] [Indexed: 09/04/2024]
Abstract
Human ALDH comprise 19 subfamilies in which ALDH1A1, ALDH1A3, ALDH3A1, ALDH5A1, ALDH7A1, and ALDH18A1 are implicated in CSC. Studies have shown that ALDH can also be involved in drug resistance and standard chemotherapy regimens are ineffective in treating patients at the stage of disease recurrence. Existing chemotherapeutic drugs eliminate the bulk of tumors but are usually not effective against CSC which express ALDH+ population. Henceforth, targeting ALDH is convincing to treat the patient's post-relapse. Combination therapies that interlink signaling mechanisms seem promising to increase the overall disease-free survival rate. Therefore, targeting ALDH through ALDH inhibitors along with immunotherapies may create a novel platform for translational research. This review aims to fill in the gap between ALDH1 family members in relation to its cell signaling mechanisms, highlighting their potential as molecular targets to sensitize recurrent tumors and bring forward the future development concerning the current progress and draw backs. This review summarizes the role of cancer stem cells and their upregulation by maintaining the tumor microenvironment in which ALDH is specifically highlighted. It discusses the regulation of ALDH family proteins and the crosstalk between ALDH and CSC in relation to cancer metabolism. Furthermore, it establishes the correlation between ALDH involved signaling mechanisms and their specific targeted inhibitors, as well as their functional modularity, bioavailability, and mechanistic role in various cancers.
Collapse
Affiliation(s)
- Kousalya Lavudi
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, United States; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, United States
| | - Shreya Madhav Nuguri
- Department of Food science and Technology, The Ohio State University, Columbus, OH, United States
| | - Prashant Pandey
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow 226025, U.P., India; Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2E1, Canada
| | | | - Qi-En Wang
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, OH 43210, United States; Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
5
|
Datta N, Vp S, Parvathy K, A S S, Maliekal TT. ALDH1A1 as a marker for metastasis initiating cells: A mechanistic insight. Exp Cell Res 2024; 442:114213. [PMID: 39173941 DOI: 10.1016/j.yexcr.2024.114213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/24/2024]
Abstract
Since metastasis accounts for the majority of cancer morbidity and mortality, attempts are focused to block metastasis and metastasis initiating cellular programs. It is generally believed that hypoxia, reactive oxygen species (ROS) and the dysregulated redox pathways regulate metastasis. Although induction of epithelial to mesenchymal transition (EMT) can initiate cell motility to different sites other than the primary site, the initiation of a secondary tumor at a distant site depends on self-renewal property of cancer stem cell (CSC) property. That subset of metastatic cells possessing CSC property are referred to as metastasis initiating cells (MICs). Among the different cellular intermediates regulating metastasis in response to hypoxia by inducing EMT and self-renewal property, ALDH1A1 is a critical molecule, which can be used as a marker for MICs in a wide variety of malignancies. The cytosolic ALDHs can irreversibly convert retinal to retinoic acid (RA), which initiates RA signaling, important for self-renewal and EMT. The metastasis permissive tumor microenvironment increases the expression of ALDH1A1, primarily through HIF1α, and leads to metabolic reprograming through OXPHOS regulation. The ALDH1A1 expression and its high activity can reprogram the cancer cells with the transcriptional upregulation of several genes, involved in EMT through RA signaling to manifest hybrid EMT or Hybrid E/M phenotype, which is important for acquiring the characteristics of MICs. Thus, the review on this topic highlights the use of ALDH1A1 as a marker for MICs, and reporters for the marker can be effectively used to trace the population in mouse models, and to screen drugs that target MICs.
Collapse
Affiliation(s)
- Nandini Datta
- Cancer Research, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala, 695014, India
| | - Snijesh Vp
- Division of Molecular Medicine, St. John's Research Institute, St John's National Academy of Health Sciences, Bangalore, 560034, India
| | - K Parvathy
- Cancer Research, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala, 695014, India
| | - Sneha A S
- Cancer Research, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala, 695014, India
| | - Tessy Thomas Maliekal
- Cancer Research, Rajiv Gandhi Centre for Biotechnology (BRIC-RGCB), Thiruvananthapuram, Kerala, 695014, India; Regional Centre for Biotechnology, Faridabad, Haryana 121001, India.
| |
Collapse
|
6
|
Chu X, Tian W, Ning J, Xiao G, Zhou Y, Wang Z, Zhai Z, Tanzhu G, Yang J, Zhou R. Cancer stem cells: advances in knowledge and implications for cancer therapy. Signal Transduct Target Ther 2024; 9:170. [PMID: 38965243 PMCID: PMC11224386 DOI: 10.1038/s41392-024-01851-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: 10/02/2023] [Revised: 03/27/2024] [Accepted: 04/28/2024] [Indexed: 07/06/2024] Open
Abstract
Cancer stem cells (CSCs), a small subset of cells in tumors that are characterized by self-renewal and continuous proliferation, lead to tumorigenesis, metastasis, and maintain tumor heterogeneity. Cancer continues to be a significant global disease burden. In the past, surgery, radiotherapy, and chemotherapy were the main cancer treatments. The technology of cancer treatments continues to develop and advance, and the emergence of targeted therapy, and immunotherapy provides more options for patients to a certain extent. However, the limitations of efficacy and treatment resistance are still inevitable. Our review begins with a brief introduction of the historical discoveries, original hypotheses, and pathways that regulate CSCs, such as WNT/β-Catenin, hedgehog, Notch, NF-κB, JAK/STAT, TGF-β, PI3K/AKT, PPAR pathway, and their crosstalk. We focus on the role of CSCs in various therapeutic outcomes and resistance, including how the treatments affect the content of CSCs and the alteration of related molecules, CSCs-mediated therapeutic resistance, and the clinical value of targeting CSCs in patients with refractory, progressed or advanced tumors. In summary, CSCs affect therapeutic efficacy, and the treatment method of targeting CSCs is still difficult to determine. Clarifying regulatory mechanisms and targeting biomarkers of CSCs is currently the mainstream idea.
Collapse
Affiliation(s)
- Xianjing Chu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wentao Tian
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jiaoyang Ning
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Gang Xiao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yunqi Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ziqi Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhuofan Zhai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Guilong Tanzhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Jie Yang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China.
| |
Collapse
|
7
|
Gowda K, Raza A, Vangala V, Lone NA, Lin JM, Singh JK, Srivastava SK, Schell TD, Robertson GP, Amin S, Sharma AK. Identification of Novel Isatin Derivative Bearing a Nitrofuran Moiety as Potent Multi-Isoform Aldehyde Dehydrogenase Inhibitor. Molecules 2024; 29:3114. [PMID: 38999066 PMCID: PMC11243058 DOI: 10.3390/molecules29133114] [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: 05/25/2024] [Revised: 06/23/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
Aldehyde dehydrogenases (ALDHs) are a family of enzymes that aid in detoxification and are overexpressed in several different malignancies. There is a correlation between increased expression of ALDH and a poor prognosis, stemness, and resistance to several drugs. Several ALDH inhibitors have been generated due to the crucial role that ALDH plays in cancer stem cells. All of these inhibitors, however, are either ineffective, very toxic, or have yet to be subjected to rigorous testing on their effectiveness. Although various drug-like compounds targeting ALDH have been reported in the literature, none have made it to routine use in the oncology clinic. As a result, new potent, non-toxic, bioavailable, and therapeutically effective ALDH inhibitors are still needed. In this study, we designed and synthesized potent multi-ALDH isoform inhibitors based on the isatin and indazole pharmacophore. Molecular docking studies and enzymatic tests revealed that among all of the synthesized analogs, compound 3 is the most potent inhibitor of ALDH1A1, ALDH3A1, and ALDH1A3, exhibiting 51.32%, 51.87%, and 36.65% inhibition, respectively. The ALDEFLUOR assay further revealed that compound 3 acts as an ALDH broad spectrum inhibitor at 500 nM. Compound 3 was also the most cytotoxic to cancer cells, with an IC50 in the range of 2.1 to 3.8 µM for ovarian, colon, and pancreatic cancer cells, compared to normal and embryonic kidney cells (IC50 7.1 to 8.7 µM). Mechanistically, compound 3 increased ROS activity due to potent multi-ALDH isoform inhibition, which increased apoptosis. Taken together, this study identified a potent multi-isoform ALDH inhibitor that could be further developed as a cancer therapeutic.
Collapse
Affiliation(s)
- Krishne Gowda
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Asif Raza
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Venugopal Vangala
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Nazir Ahmad Lone
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Jyh Ming Lin
- Department of Biochemistry and Molecular Biology, Penn State Cancer Institute, Penn State College of Medicine Hershey, Hershey, PA 17033, USA
| | - Jaikee Kumar Singh
- Department of Biosciences, Manipal University Jaipur, Jaipur 303007, India (S.K.S.)
| | | | - Todd D. Schell
- Department of Microbiology and Immunology, Penn State Cancer Institute, Penn State College of Medicine Hershey, Hershey, PA 17033, USA
| | - Gavin P. Robertson
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA 17033, USA
- Departments of Pathology, Dermatology, Surgery, Melanoma Skin Cancer Center, Penn State Cancer Institute, Penn State College of Medicine Hershey, Hershey, PA 17033, USA
| | - Shantu Amin
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Arun K. Sharma
- Department of Pharmacology, Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
8
|
Temizer AB, Uludoğan G, Özçelik R, Koulani T, Ozkirimli E, Ulgen KO, Karali N, Özgür A. Exploring data-driven chemical SMILES tokenization approaches to identify key protein-ligand binding moieties. Mol Inform 2024; 43:e202300249. [PMID: 38196065 DOI: 10.1002/minf.202300249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/13/2023] [Accepted: 01/06/2024] [Indexed: 01/11/2024]
Abstract
Machine learning models have found numerous successful applications in computational drug discovery. A large body of these models represents molecules as sequences since molecular sequences are easily available, simple, and informative. The sequence-based models often segment molecular sequences into pieces called chemical words, analogous to the words that make up sentences in human languages, and then apply advanced natural language processing techniques for tasks such as de novo drug design, property prediction, and binding affinity prediction. However, the chemical characteristics and significance of these building blocks, chemical words, remain unexplored. To address this gap, we employ data-driven SMILES tokenization techniques such as Byte Pair Encoding, WordPiece, and Unigram to identify chemical words and compare the resulting vocabularies. To understand the chemical significance of these words, we build a language-inspired pipeline that treats high affinity ligands of protein targets as documents and selects key chemical words making up those ligands based on tf-idf weighting. The experiments on multiple protein-ligand affinity datasets show that despite differences in words, lengths, and validity among the vocabularies generated by different subword tokenization algorithms, the identified key chemical words exhibit similarity. Further, we conduct case studies on a number of target to analyze the impact of key chemical words on binding. We find that these key chemical words are specific to protein targets and correspond to known pharmacophores and functional groups. Our approach elucidates chemical properties of the words identified by machine learning models and can be used in drug discovery studies to determine significant chemical moieties.
Collapse
Affiliation(s)
- Asu Busra Temizer
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, İstanbul University, İstanbul, Turkey
- Department of Pharmaceutical Chemistry, Institute of Health Sciences, İstanbul University, İstanbul, Turkey
| | - Gökçe Uludoğan
- Department of Computer Engineering, Boğaziçi University, İstanbul, Turkey
| | - Rıza Özçelik
- Department of Computer Engineering, Boğaziçi University, İstanbul, Turkey
| | - Taha Koulani
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, İstanbul University, İstanbul, Turkey
- Department of Pharmaceutical Chemistry, Institute of Health Sciences, İstanbul University, İstanbul, Turkey
| | - Elif Ozkirimli
- Science and Research Informatics, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Kutlu O Ulgen
- Department of Chemical Engineering, Boğaziçi University, İstanbul, Turkey
| | - Nilgun Karali
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, İstanbul University, İstanbul, Turkey
| | - Arzucan Özgür
- Department of Computer Engineering, Boğaziçi University, İstanbul, Turkey
| |
Collapse
|
9
|
Singh AK, Prasad P, Cancelas JA. Mesenchymal stromal cells, metabolism, and mitochondrial transfer in bone marrow normal and malignant hematopoiesis. Front Cell Dev Biol 2023; 11:1325291. [PMID: 38169927 PMCID: PMC10759248 DOI: 10.3389/fcell.2023.1325291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
Hematopoietic stem cell (HSC) transplantation-based treatments are in different phases of clinical development, ranging from current therapies to a promise in the repair and regeneration of diseased tissues and organs. Mesenchymal stromal/stem cells (MSCs), which are fibroblast-like heterogeneous progenitors with multilineage differentiation (osteogenic, chondrogenic, and adipogenic) and self-renewal potential, and exist in the bone marrow (BM), adipose, and synovium, among other tissues, represent one of the most widely used sources of stem cells in regenerative medicine. MSCs derived from bone marrow (BM-MSCs) exhibit a variety of traits, including the potential to drive HSC fate and anti-inflammatory and immunosuppressive capabilities via paracrine activities and interactions with the innate and adaptive immune systems. The role of BM-MSC-derived adipocytes is more controversial and may act as positive or negative regulators of benign or malignant hematopoiesis based on their anatomical location and functional crosstalk with surrounding cells in the BM microenvironment. This review highlights the most recent clinical and pre-clinical findings on how BM-MSCs interact with the surrounding HSCs, progenitors, and immune cells, and address some recent insights on the mechanisms that mediate MSCs and adipocyte metabolic control through a metabolic crosstalk between BM microenvironment cells and intercellular mitochondrial transfer in normal and malignant hematopoiesis.
Collapse
Affiliation(s)
- Abhishek K. Singh
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Parash Prasad
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Jose A. Cancelas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
10
|
Huang G, Hucek D, Cierpicki T, Grembecka J. Applications of oxetanes in drug discovery and medicinal chemistry. Eur J Med Chem 2023; 261:115802. [PMID: 37713805 DOI: 10.1016/j.ejmech.2023.115802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/17/2023]
Abstract
The compact and versatile oxetane motifs have gained significant attention in drug discovery and medicinal chemistry campaigns. This review presents an overview of the diverse applications of oxetanes in clinical and preclinical drug candidates targeting various human diseases, including cancer, viral infections, autoimmune disorders, neurodegenerative conditions, metabolic disorders, and others. Special attention is given to biologically active oxetane-containing compounds and their disease-related targets, such as kinases, epigenetic and non-epigenetic enzymes, and receptors. The review also details the effect of the oxetane motif on important properties, including aqueous solubility, lipophilicity, pKa, P-glycoprotein (P-gp) efflux, metabolic stability, conformational preferences, toxicity profiles (e.g., cytochrome P450 (CYP) suppression and human ether-a-go-go related gene (hERG) inhibition), pharmacokinetic (PK) properties, potency, and target selectivity. We anticipate that this work will provide valuable insights that can drive future discoveries of novel bioactive oxetane-containing small molecules, enabling their effective application in combating a wide range of human diseases.
Collapse
Affiliation(s)
- Guang Huang
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Devon Hucek
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Tomasz Cierpicki
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jolanta Grembecka
- Department of Pathology, University of Michigan, Ann Arbor, MI, 48109, USA
| |
Collapse
|
11
|
Hazrati A, Malekpour K, Mirsanei Z, Khosrojerdi A, Rahmani-Kukia N, Heidari N, Abbasi A, Soudi S. Cancer-associated mesenchymal stem/stromal cells: role in progression and potential targets for therapeutic approaches. Front Immunol 2023; 14:1280601. [PMID: 38022534 PMCID: PMC10655012 DOI: 10.3389/fimmu.2023.1280601] [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: 08/20/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023] Open
Abstract
Malignancies contain a relatively small number of Mesenchymal stem/stromal cells (MSCs), constituting a crucial tumor microenvironment (TME) component. These cells comprise approximately 0.01-5% of the total TME cell population. MSC differentiation potential and their interaction with the tumor environment enable these cells to affect tumor cells' growth, immune evasion, metastasis, drug resistance, and angiogenesis. This type of MSC, known as cancer-associated mesenchymal stem/stromal cells (CA-MSCs (interacts with tumor/non-tumor cells in the TME and affects their function by producing cytokines, chemokines, and various growth factors to facilitate tumor cell migration, survival, proliferation, and tumor progression. Considering that the effect of different cells on each other in the TME is a multi-faceted relationship, it is essential to discover the role of these relationships for targeting in tumor therapy. Due to the immunomodulatory role and the tissue repair characteristic of MSCs, these cells can help tumor growth from different aspects. CA-MSCs indirectly suppress antitumor immune response through several mechanisms, including decreasing dendritic cells (DCs) antigen presentation potential, disrupting natural killer (NK) cell differentiation, inducing immunoinhibitory subsets like tumor-associated macrophages (TAMs) and Treg cells, and immune checkpoint expression to reduce effector T cell antitumor responses. Therefore, if these cells can be targeted for treatment so that their population decreases, we can hope for the treatment and improvement of the tumor conditions. Also, various studies show that CA-MSCs in the TME can affect other vital aspects of a tumor, including cell proliferation, drug resistance, angiogenesis, and tumor cell invasion and metastasis. In this review article, we will discuss in detail some of the mechanisms by which CA-MSCs suppress the innate and adaptive immune systems and other mechanisms related to tumor progression.
Collapse
Affiliation(s)
- Ali Hazrati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Kosar Malekpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Mirsanei
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arezou Khosrojerdi
- Infectious Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Nasim Rahmani-Kukia
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Neda Heidari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ardeshir Abbasi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Sara Soudi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
12
|
Manouchehri JM, Marcho L, Cherian MA. Sulfatase 2 Inhibition Sensitizes Triple-Negative Breast Cancer Cells to Chemotherapy Through Augmentation of Extracellular ATP. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.15.557965. [PMID: 37745565 PMCID: PMC10516004 DOI: 10.1101/2023.09.15.557965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Background Breast cancer is the leading cause of cancer-related death among women worldwide. Patients diagnosed with triple-negative breast cancer (TNBC) have limited therapeutic options that produce durable responses. Hence, a diagnosis of TNBC is associated with a poor prognosis compared to other types of breast cancer. As a result, there is a critical need for novel therapies that can deepen and prolong responses.We previously found that chemotherapy causes the release of extracellular adenosine triphosphate (eATP). Augmenting eATP release can boost the response of TNBC cells to chemotherapy and cause increased cell death. However, eATP concentrations are limited by several families of extracellular ATPases, which complicates the design of compounds that attenuate eATP degradation.In this study, we hypothesized that heparan sulfate (HS) would inhibit extracellular ATPases and accentuate chemotherapy-induced cytotoxicity in TNBC by augmenting eATP. HS can be desulfated by sulfatase 1 and 2; sulfatase 2 is consistently highly expressed in a variety of cancers including breast cancer, whereas sulfatase 1 is not. We hypothesized that the sulfatase 2 inhibitor OKN-007 would exacerbate chemotherapy-induced eATP release and TNBC cell death. Methods TNBC cell lines and nontumorigenic immortal mammary epithelial cells were treated with paclitaxel in the presence of heparan sodium sulfate and/or OKN-007; eATP content and cell viability were evaluated. In addition, protein and cell surface expression of sulfatases 1 and 2 were determined in all examined cell lines via ELISA, Western blot, and flow cytometry analyses. Results Sulfatase 2 was highly expressed in TNBC cell lines and human breast cancer samples but not in immortal mammary epithelial cells and much less so in normal human breast tissue and ductal carcinoma in situ samples. OKN-007 exacerbated chemotherapy-induced eATP release and chemotherapy-induced TNBC cell death. When combined with chemotherapy, OKN-007 attenuated cells with a cancer-initiating cell phenotype. Conclusions These results suggest that sulfatase 2 inhibitors in combination with chemotherapy attenuate the viability of TNBC cells more than chemotherapy alone by exacerbating eATP release. These effects, as well as their capacity to attenuate the cancer-initiating cell fraction, may translate into combination therapies for TNBC that induce deeper and more durable responses.
Collapse
|
13
|
Manouchehri JM, Marcho L, Cherian MA. The role of heparan sulfate in enhancing the chemotherapeutic response in triple-negative breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.08.556819. [PMID: 37745355 PMCID: PMC10515779 DOI: 10.1101/2023.09.08.556819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Background Among women worldwide, breast cancer has the highest incidence and is the leading cause of cancer-related death. Patients with the triple-negative breast cancer (TNBC) subtype have an inferior prognosis in comparison to other breast cancers because current therapies do not facilitate long-lasting responses. Thus, there is a demand for more innovative therapies that induce durable responses.In our previous research, we discovered that augmenting the concentration of extracellular ATP (eATP) greatly enhances the chemotherapeutic response of TNBC cell lines by activating purinergic receptors (P2RXs), leading to cell death through the induction of non-selective membrane permeability. However, eATP levels are limited by several classes of extracellular ATPases. One endogenous molecule of interest that can inhibit multiple classes of extracellular ATPases is heparan sulfate. Polysulfated polysaccharide heparan sulfate itself is degraded by heparanase, an enzyme that is known to be highly expressed in various cancers, including breast cancer. Heparan sulfate has previously been shown to regulate several cancer-related processes such as fibroblast growth factor signaling, neoangiogenesis by sequestering vascular endothelial growth factors in the extracellular matrix, hedgehog signaling and cell adhesion. In this project, we identified an additional mechanism for a tumor suppressor role of heparan sulfate: inhibition of extracellular ATPases, leading to augmented levels of eATP.Several heparanase inhibitors have been previously identified, including OGT 2115, suramin, PI-88, and PG 545. We hypothesized that heparanase inhibitors would augment eATP concentrations in TNBC by increasing heparan sulfate in the tumor microenvironment, resulting in enhanced cell death in response to chemotherapy. Methods We treated TNBC cell lines MDA-MB 231, Hs 578t, and MDA-MB 468 and non-tumorigenic immortal mammary epithelial MCF-10A cells with increasing concentrations of the chemotherapeutic agent paclitaxel in the presence of heparan sulfate and/or the heparanase inhibitor OGT 2115 while analyzing eATP release and cell viability. Moreover, to verify that the effects of OGT 2115 are mediated through eATP, we applied specific antagonists to the purinergic receptors P2RX4 and P2RX7. In addition, the protein expression of heparanase was compared in the cell lines by Western blot analysis. We also evaluated the consequences of this therapeutic strategy on the breast cancer-initiating cell population in the treated cells using flow cytometry and tumorsphere formation efficiency assays. Results Heparanase was found to be highly expressed in immortal mammary epithelial cells in comparison to TNBC cell lines. The heparanase inhibitor OGT 2115 augmented chemotherapy-induced TNBC cell death and eATP release. Conclusion These results demonstrate that inhibiting the degradation of heparan sulfate in the tumor microenvironment augments the susceptibility of TNBC cell lines to chemotherapy by increasing extracellular ATP concentrations. This strategy could potentially be applied to induce more enhanced and enduring responses in TNBC patients.
Collapse
|
14
|
Xanthis V, Mantso T, Dimtsi A, Pappa A, Fadouloglou VE. Human Aldehyde Dehydrogenases: A Superfamily of Similar Yet Different Proteins Highly Related to Cancer. Cancers (Basel) 2023; 15:4419. [PMID: 37686694 PMCID: PMC10650815 DOI: 10.3390/cancers15174419] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
The superfamily of human aldehyde dehydrogenases (hALDHs) consists of 19 isoenzymes which are critical for several physiological and biosynthetic processes and play a major role in the organism's detoxification via the NAD(P) dependent oxidation of numerous endogenous and exogenous aldehyde substrates to their corresponding carboxylic acids. Over the last decades, ALDHs have been the subject of several studies as it was revealed that their differential expression patterns in various cancer types are associated either with carcinogenesis or promotion of cell survival. Here, we attempt to provide a thorough review of hALDHs' diverse functions and 3D structures with particular emphasis on their role in cancer pathology and resistance to chemotherapy. We are especially interested in findings regarding the association of structural features and their changes with effects on enzymes' functionalities. Moreover, we provide an updated outline of the hALDHs inhibitors utilized in experimental or clinical settings for cancer therapy. Overall, this review aims to provide a better understanding of the impact of ALDHs in cancer pathology and therapy from a structural perspective.
Collapse
Affiliation(s)
| | | | | | | | - Vasiliki E. Fadouloglou
- Department of Molecular Biology & Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
15
|
Zhao X, Guo B, Sun W, Yu J, Cui L. Targeting Squalene Epoxidase Confers Metabolic Vulnerability and Overcomes Chemoresistance in HNSCC. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206878. [PMID: 37490552 PMCID: PMC10520660 DOI: 10.1002/advs.202206878] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 06/08/2023] [Indexed: 07/27/2023]
Abstract
Cisplatin resistance poses a substantial hurdle in effectively treating head and neck squamous cell carcinoma (HNSCC). Utilizing multiple tumor models and examining an internal HNSCC cohort, squalene epoxidase (SQLE) is pinpointed as a key driver of chemoresistance and tumorigenesis, operating through a cholesterol-dependent pathway. Comprehensive transcriptomic analysis reveals that SQLE is essential for maintaining c-Myc transcriptional activity by stabilizing the c-Myc protein and averting its ubiquitin-mediated degradation. Mechanistic investigation demonstrates that SQLE inhibition diminishes Akt's binding affinity to lipid rafts via a cholesterol-dependent process, subsequently deactivating lipid raft-localized Akt, reducing GSK-3β phosphorylation at S9, and increasing c-Myc phosphorylation at T58, ultimately leading to c-Myc destabilization. Importantly, employing an Sqle conditional knockout mouse model, SQLE's critical role in HNSCC initiation and progression is established. The preclinical findings demonstrate the potent synergistic effects of combining terbinafine and cisplatin in arresting tumor growth. These discoveries not only provide novel insights into the underlying mechanisms of SQLE-mediated cisplatin resistance and tumorigenesis in HNSCC but also propose a promising therapeutic avenue for HNSCC patients unresponsive to conventional cisplatin-based chemotherapy.
Collapse
Affiliation(s)
- Xinyuan Zhao
- Stomatological HospitalSchool of StomatologySouthern Medical UniversityGuangzhou510280China
| | - Bing Guo
- Department of Dentistrythe First Affiliated HospitalSun Yat‐sen UniversityGuangzhou510080China
| | - Wenjuan Sun
- Department of StomatologyThe Third Affiliated HospitalSun Yat‐sen UniversityGuangzhou510630China
| | - Jinhua Yu
- Department of EndodonticsJiangsu Key Laboratory of Oral DiseasesAffiliated Hospital of StomatologyNanjing Medical UniversityNanjing210029China
| | - Li Cui
- Stomatological HospitalSchool of StomatologySouthern Medical UniversityGuangzhou510280China
- Division of Oral Biology and MedicineSchool of DentistryUniversity of California Los AngelesLos AngelesCA90095USA
| |
Collapse
|
16
|
Świerczewska M, Sterzyńska K, Ruciński M, Andrzejewska M, Nowicki M, Januchowski R. The response and resistance to drugs in ovarian cancer cell lines in 2D monolayers and 3D spheroids. Biomed Pharmacother 2023; 165:115152. [PMID: 37442067 DOI: 10.1016/j.biopha.2023.115152] [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: 05/16/2023] [Revised: 07/06/2023] [Accepted: 07/07/2023] [Indexed: 07/15/2023] Open
Abstract
Ovarian cancer is the most common type of gynecologic cancer. One of the leading causes of high mortality is chemoresistance, developed primarily or during treatment. Different mechanisms of drug resistance appear at the cellular and cancer tissue organization levels. We examined the differences in response to the cytotoxic drugs CIS, MTX, DOX, VIN, PAC, and TOP using 2D (two-dimensional) and 3D (three-dimensional) culture methods. We tested the drug-sensitive ovarian cancer cell line W1 and established resistant cell lines to appropriate cytotoxic drugs. The following qualitative and quantitative methods were used to assess: 1) morphology - inverted microscope and hematoxylin & eosin staining; 2) viability - MTT assay; 3) gene expression - a quantitative polymerase chain reaction; 4) identification of proteins - immunohistochemistry, and immunofluorescence. Our results indicate that the drug-sensitive and drug-resistant cells cultured in 3D conditions exhibit stronger resistance than the cells cultured in 2D conditions. A traditional 2D model shows that drug resistance of cancer cells is caused mainly by changes in the expression of genes encoding ATP-binding cassette transporter proteins, components of the extracellular matrix, "new" established genes related to drug resistance in ovarian cancer cell lines, and universal marker of cancer stem cells. Whereas in a 3D model, the drug resistance in spheroids can be related to other mechanisms such as the structure of the spheroid (dense or loose), the cell type (necrotic, quiescent, proliferating cells), drug concentrations or drug diffusion into the dense cellular/ECM structure.
Collapse
Affiliation(s)
- Monika Świerczewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland.
| | - Karolina Sterzyńska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland.
| | - Marcin Ruciński
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland.
| | - Małgorzata Andrzejewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland.
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznan, Poland.
| | - Radosław Januchowski
- Institute of Health Sciences, Collegium Medicum, University of Zielona Góra, Zyty 28 St., 65-046 Zielona Góra, Poland.
| |
Collapse
|
17
|
Gomez-Salazar MA, Wang Y, Thottappillil N, Hardy RW, Alexandre M, Höller F, Martin N, Gonzalez-Galofre ZN, Stefancova D, Medici D, James AW, Péault B. Aldehyde Dehydrogenase, a Marker of Normal and Malignant Stem Cells, Typifies Mesenchymal Progenitors in Perivascular Niches. Stem Cells Transl Med 2023; 12:474-484. [PMID: 37261440 PMCID: PMC10651226 DOI: 10.1093/stcltm/szad024] [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: 06/22/2022] [Accepted: 04/07/2023] [Indexed: 06/02/2023] Open
Abstract
Innate mesenchymal stem cells exhibiting multilineage differentiation and tissue (re)generative-or pathogenic-properties reside in perivascular niches. Subsets of these progenitors are committed to either osteo-, adipo-, or fibrogenesis, suggesting the existence of a developmental organization in blood vessel walls. We evaluated herein the activity of aldehyde dehydrogenase, a family of enzymes catalyzing the oxidation of aldehydes into carboxylic acids and a reported biomarker of normal and malignant stem cells, within human adipose tissue perivascular areas. A progression of ALDHLow to ALDHHigh CD34+ cells was identified in the tunica adventitia. Mesenchymal stem cell potential was confined to ALDHHigh cells, as assessed by proliferation and multilineage differentiation in vitro of cells sorted by flow cytometry with a fluorescent ALDH substrate. RNA sequencing confirmed and validated that ALDHHigh cells have a progenitor cell phenotype and provided evidence that the main isoform in this fraction is ALDH1A1, which was confirmed by immunohistochemistry. This demonstrates that ALDH activity, which marks hematopoietic progenitors and stem cells in diverse malignant tumors, also typifies native, blood vessel resident mesenchymal stem cells.
Collapse
Affiliation(s)
- Mario A Gomez-Salazar
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
- Department of Pathology, Johns Hopkins University, Baltimore, MB, USA
| | - Yiyun Wang
- Department of Pathology, Johns Hopkins University, Baltimore, MB, USA
| | | | - Reef W Hardy
- Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Manon Alexandre
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
- Polytech Marseille, Aix Marseille University, Marseille, France
| | - Fabian Höller
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Niall Martin
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Zaniah N Gonzalez-Galofre
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Dorota Stefancova
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Daniele Medici
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
| | - Aaron W James
- Department of Pathology, Johns Hopkins University, Baltimore, MB, USA
| | - Bruno Péault
- Center for Regenerative Medicine and Center for Cardiovascular Research, University of Edinburgh, Edinburgh, UK
- Orthopaedic Hospital Research Center and Broad Stem Cell Research Center, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| |
Collapse
|
18
|
Tóthová Z, Šemeláková M, Bhide K, Bhide M, Kováč A, Majerová P, Kvaková M, Štofilová J, Solárová Z, Solár P. Differentially Expressed Genes Induced by Erythropoietin Receptor Overexpression in Rat Mammary Adenocarcinoma RAMA 37-28 Cells. Int J Mol Sci 2023; 24:ijms24108482. [PMID: 37239828 DOI: 10.3390/ijms24108482] [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: 03/29/2023] [Revised: 04/25/2023] [Accepted: 05/02/2023] [Indexed: 05/28/2023] Open
Abstract
The erythropoietin receptor (EPOR) is a transmembrane type I receptor with an essential role in the proliferation and differentiation of erythroid progenitors. Besides its function during erythropoiesis, EPOR is expressed and has protective effect in various non-hematopoietic tissues, including tumors. Currently, the advantageous aspect of EPOR related to different cellular events is still under scientific investigation. Besides its well-known effect on cell proliferation, apoptosis and differentiation, our integrative functional study revealed its possible associations with metabolic processes, transport of small molecules, signal transduction and tumorigenesis. Comparative transcriptome analysis (RNA-seq) identified 233 differentially expressed genes (DEGs) in EPOR overexpressed RAMA 37-28 cells compared to parental RAMA 37 cells, whereas 145 genes were downregulated and 88 upregulated. Of these, for example, GPC4, RAP2C, STK26, ZFP955A, KIT, GAS6, PTPRF and CXCR4 were downregulated and CDH13, NR0B1, OCM2, GPM6B, TM7SF3, PARVB, VEGFD and STAT5A were upregulated. Surprisingly, two ephrin receptors, EPHA4 and EPHB3, and EFNB1 ligand were found to be upregulated as well. Our study is the first demonstrating robust differentially expressed genes evoked by simple EPOR overexpression without the addition of erythropoietin ligand in a manner which remains to be elucidated.
Collapse
Affiliation(s)
- Zuzana Tóthová
- Department of Medical Biology, Faculty of Medicine, P.J. Šafárik University in Košice, 04001 Košice, Slovakia
| | - Martina Šemeláková
- Department of Medical Biology, Faculty of Medicine, P.J. Šafárik University in Košice, 04001 Košice, Slovakia
| | - Katarína Bhide
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Košice, 04001 Košice, Slovakia
| | - Mangesh Bhide
- Laboratory of Biomedical Microbiology and Immunology, University of Veterinary Medicine and Pharmacy in Košice, 04001 Košice, Slovakia
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Andrej Kováč
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Petra Majerová
- Institute of Neuroimmunology, Slovak Academy of Sciences, 84510 Bratislava, Slovakia
| | - Monika Kvaková
- Department of Experimental Medicine, Faculty of Medicine, P.J. Šafárik University in Košice, 04001 Košice, Slovakia
| | - Jana Štofilová
- Department of Experimental Medicine, Faculty of Medicine, P.J. Šafárik University in Košice, 04001 Košice, Slovakia
| | - Zuzana Solárová
- Department of Pharmacology, Faculty of Medicine, P.J. Šafárik University in Košice, 04001 Košice, Slovakia
| | - Peter Solár
- Department of Medical Biology, Faculty of Medicine, P.J. Šafárik University in Košice, 04001 Košice, Slovakia
| |
Collapse
|
19
|
Microbiota-Derived Natural Products Targeting Cancer Stem Cells: Inside the Gut Pharma Factory. Int J Mol Sci 2023; 24:ijms24054997. [PMID: 36902427 PMCID: PMC10003410 DOI: 10.3390/ijms24054997] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Cancer stem cells (CSCs) have drawn much attention as important tumour-initiating cells that may also be crucial for recurrence after chemotherapy. Although the activity of CSCs in various forms of cancer is complex and yet to be fully elucidated, opportunities for therapies targeting CSCs exist. CSCs are molecularly distinct from bulk tumour cells, so they can be targeted by exploiting their signature molecular pathways. Inhibiting stemness has the potential to reduce the risk posed by CSCs by limiting or eliminating their capacity for tumorigenesis, proliferation, metastasis, and recurrence. Here, we briefly described the role of CSCs in tumour biology, the mechanisms involved in CSC therapy resistance, and the role of the gut microbiota in cancer development and treatment, to then review and discuss the current advances in the discovery of microbiota-derived natural compounds targeting CSCs. Collectively, our overview suggests that dietary intervention, toward the production of those identified microbial metabolites capable of suppressing CSC properties, is a promising approach to support standard chemotherapy.
Collapse
|
20
|
Al-Shamma SA, Zaher DM, Hersi F, Abu Jayab NN, Omar HA. Targeting aldehyde dehydrogenase enzymes in combination with chemotherapy and immunotherapy: An approach to tackle resistance in cancer cells. Life Sci 2023; 320:121541. [PMID: 36870386 DOI: 10.1016/j.lfs.2023.121541] [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: 12/19/2022] [Revised: 02/19/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023]
Abstract
Modern cancer chemotherapy originated in the 1940s, and since then, many chemotherapeutic agents have been developed. However, most of these agents show limited response in patients due to innate and acquired resistance to therapy, which leads to the development of multi-drug resistance to different treatment modalities, leading to cancer recurrence and, eventually, patient death. One of the crucial players in inducing chemotherapy resistance is the aldehyde dehydrogenase (ALDH) enzyme. ALDH is overexpressed in chemotherapy-resistant cancer cells, which detoxifies the generated toxic aldehydes from chemotherapy, preventing the formation of reactive oxygen species and, thus, inhibiting the induction of oxidative stress and the stimulation of DNA damage and cell death. This review discusses the mechanisms of chemotherapy resistance in cancer cells promoted by ALDH. In addition, we provide detailed insight into the role of ALDH in cancer stemness, metastasis, metabolism, and cell death. Several studies investigated targeting ALDH in combination with other treatments as a potential therapeutic regimen to overcome resistance. We also highlight novel approaches in ALDH inhibition, including the potential synergistic employment of ALDH inhibitors in combination with chemotherapy or immunotherapy against different cancers, including head and neck, colorectal, breast, lung, and liver.
Collapse
Affiliation(s)
- Salma A Al-Shamma
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Dana M Zaher
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Fatema Hersi
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Nour N Abu Jayab
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hany A Omar
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| |
Collapse
|
21
|
Identity matters: cancer stem cells and tumour plasticity in head and neck squamous cell carcinoma. Expert Rev Mol Med 2023; 25:e8. [PMID: 36740973 DOI: 10.1017/erm.2023.4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Head and neck squamous cell carcinoma (HNSCC) represents frequent yet aggressive tumours that encompass complex ecosystems of stromal and neoplastic components including a dynamic population of cancer stem cells (CSCs). Recently, research in the field of CSCs has gained increased momentum owing in part to their role in tumourigenicity, metastasis, therapy resistance and relapse. We provide herein a comprehensive assessment of the latest progress in comprehending CSC plasticity, including newly discovered influencing factors and their possible application in HNSCC. We further discuss the dynamic interplay of CSCs within tumour microenvironment considering our evolving appreciation of the contribution of oral microbiota and the pressing need for relevant models depicting their features. In sum, CSCs and tumour plasticity represent an exciting and expanding battleground with great implications for cancer therapy that are only beginning to be appreciated in head and neck oncology.
Collapse
|
22
|
Izycka N, Rucinski M, Andrzejewska M, Szubert S, Nowak-Markwitz E, Sterzynska K. The Prognostic Value of Cancer Stem Cell Markers (CSCs) Expression-ALDH1A1, CD133, CD44-For Survival and Long-Term Follow-Up of Ovarian Cancer Patients. Int J Mol Sci 2023; 24:ijms24032400. [PMID: 36768723 PMCID: PMC9916537 DOI: 10.3390/ijms24032400] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/18/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
Recurrent disease and treatment-associated chemoresistance are the two main factors accounting for poor clinical outcomes of ovarian cancer (OC) patients. Both can be associated with cancer stem cells (CSCs), which contribute to cancer formation, progression, chemoresistance, and recurrence. Hence, this study investigated whether the expression of known CSC-associated markers ALDH1A, CD44, and CD133 may predict OC patient prognosis. We analyzed their expression in primary epithelial ovarian cancer (EOC) patients using immunohistochemistry and related them to clinicopathological data, including overall survival (OS) and progression-free survival (PFS). Expression of ALDH1A1 was detected in 32%, CD133 in 28%, and CD44 in 33% of cases. While Kaplan-Meier analysis revealed no association of the expression of CD133 and CD44 with PFS and OS, ALDH1A1-positive patients were characterized with both significantly shorter OS (p = 0.00022) and PFS (p = 0.027). Multivariate analysis demonstrated that the expression of ALDH1A1, FIGO stage III-IV, and residual disease after suboptimal debulking or neoadjuvant chemotherapy correlated with shorter OS. The results of this study identify ALDH1A1 as a potential independent prognostic factor of shorter OS and PFS in EOC patients. Therefore, targeting ALDH1A1-positive cancer cells may be a promising therapeutic strategy to influence the disease course and treatment response.
Collapse
Affiliation(s)
- Natalia Izycka
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznań, Poland
| | - Marcin Rucinski
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland
| | - Malgorzata Andrzejewska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland
| | - Sebastian Szubert
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznań, Poland
| | - Ewa Nowak-Markwitz
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Division of Gynecologic Oncology, Poznan University of Medical Sciences, Polna 33 St., 60-535 Poznań, Poland
| | - Karolina Sterzynska
- Department of Histology and Embryology, Poznan University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland
- Correspondence: ; Tel.: +48-61-8546455
| |
Collapse
|
23
|
Wen X, Wu Y, Lou Y, Xia Y, Yu X. The roles of Linc-ROR in the regulation of cancer stem cells. Transl Oncol 2022; 28:101602. [PMID: 36535192 PMCID: PMC9791587 DOI: 10.1016/j.tranon.2022.101602] [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: 09/19/2022] [Revised: 11/06/2022] [Accepted: 12/04/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer stem cells (CSCs) are considered to be a kind of tumor cell population characterized by self-renewal, easy to metastasize and drug resistance, which play an indispensable role in the occurrence, development, metastasis and drug resistance of tumors, and their existence is an important reason for high metastasis and recurrence of tumors. Long non-coding RNAs (LncRNAs), which are more than 200 nucleotides in length, have a close relationship with the malignant progression of cancer.In recent years, abundant studies have reavling that LncRNAs are beneficial to the regulation of various cancer stem cells. Linc-ROR, as a newly discovered intergenic non-protein-coding RNA in recent years, is considered to be a key regulator affecting the development of human tumors. Dysregulation of Linc-ROR is related to stemness phenotype and functional regulation of cancer stem cells. For that, Linc-ROR has the potential to be used as a diagnostic biomarker for cancer patients and can serve as a clinically meaningful potential therapeutic target. In this review, we generalize the existing research results on the important role of Linc-ROR in regulation of CSCs.
Collapse
Affiliation(s)
- Xiaoling Wen
- Department of Gynecology, the Affiliated Hospital of Qingdao University, Qingdao, 266003,China
| | - Yingying Wu
- Department of Gynecology, the Affiliated Hospital of Qingdao University, Qingdao, 266003,China
| | - Yanhui Lou
- Department of Gynecology, the Affiliated Hospital of Qingdao University, Qingdao, 266003,China..
| | - Yufang Xia
- Department of Gynecology, the Affiliated Hospital of Qingdao University, Qingdao, 266003,China
| | - Xiao Yu
- Department of Gynecology, the Affiliated Hospital of Qingdao University, Qingdao, 266003,China
| |
Collapse
|
24
|
Wang Q, Li Z, Hao Y, Zhang Y, Zhang C. Near-Infrared Fluorescence Probe with a New Recognition Moiety for Specific Detection and Imaging of Aldehyde Dehydrogenase Expecting the Identification and Isolation of Cancer Stem Cells. Anal Chem 2022; 94:17328-17333. [PMID: 36453832 DOI: 10.1021/acs.analchem.2c04801] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Aldehyde dehydrogenase (ALDH) is a vital enzyme that converts aldehyde to acetic acid during alcohol metabolism. ALDH is also a cellular marker of cancer stem cells (CSCs), which plays an important role in cancer diagnosis and prognosis assessment. Therefore, there is a need to explore convenient, selective, and sensitive methods for the detection and imaging of ALDH. Because of the low background fluorescence and high penetration, near-infrared (NIR) fluorescent probes are powerful tools for the detection of ALDH. Until now, only one NIR fluorescent probe has been reported for detecting ALDH. Hence, we synthesized a novel NIR fluorescent probe, Probe-ALDH, by linking the new specific recognition moiety 4-hydroxymethyl benzaldehyde with NIR fluorophore AXPI. Compared with the existing ALDH fluorescent probes, Probe-ALDH has excellent properties, such as a new specific recognition moiety without the substitution of benzaldehyde, a simple synthesis method, emission wavelength in the NIR region, reaction time of only 30 min, and a detection limit as low as 0.03 U·mL-1, which is better than those of the previously reported probes. The probe effectively eliminates the interference from reactive oxygen species (ROS), amino acids, and amines. More importantly, the flow cytometry results showed that Probe-ALDH has great potential applications in the identification and isolation of CSCs. Ultimately, it was successfully applied to the imaging analysis of endogenous ALDH in HepG2 cells by the addition of inhibitor disulfiram. The excellent performance of Probe-ALDH makes it a promising candidate for drug discovery, cancer diagnosis, and so forth.
Collapse
Affiliation(s)
- Qiuyue Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Zhao Li
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Yitong Hao
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Yuan Zhang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety Control, and Shaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural Products, College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Chengxiao Zhang
- Key Laboratory of Analytical Chemistry for Life Science of Shaanxi Province, School of Chemistry and Chemical Engineering, Shaanxi Normal University, Xi'an 710062, China
| |
Collapse
|
25
|
Barrios O, Sánchez BG, Rodríguez-Prieto T, Cano J, Bort A, Gómez R, Díaz-Laviada I. Alteration of the HIF-1α/VEGF Signaling Pathway and Disruption of the Cell Cycle by Second Generation Carbosilan Dendrimers. Biomacromolecules 2022; 23:5043-5055. [PMID: 36445323 DOI: 10.1021/acs.biomac.2c00899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Current therapies against prostate cancer (PCa) disease, such as surgery, radiotherapy, or in last term chemical castration by androgen deprivation, have led to significant reduction of the incidence of PCa throughout the world. Worse prognosis is found in those patients which exhibit castration resistance, relapsing into the disease with even greater aggressiveness. Hypoxia cancer cell adaption has been observed to be closely connected to fatal prognostic tumor features. Therefore, hypoxia adaptive mechanisms of cancer cells have attracted large interest as a relevant biological target for treatment-resistant patients. Dendrimers have been established as a promising nanotechnological tool owing to their beneficial physicochemical features such as multivalency and monodispersity. Herein, we have completed a thorough study to better understand the effect within the cell of the already published ruthenium(II)-N-heterocyclic carbene metallodendrimer (G2Ru) that was able to drastically reduce HIF-1α stabilization and exhibited antiproliferative capability against androgen-sensitive (LNCaP) and androgen-resistant prostate cancer cells (LNFLU) in vitro. G2Ru, as well as its cationic imidazolium precursor (G2P), displayed scavenging properties against intracellular and externally stimulated ROS levels, which would presumably hinder the stabilization of HIF-1α by prolyl hydroxylase (PHD) inhibition. Furthermore, these dendrimers have shown considerably beneficial properties against tumor progression capability in terms of apoptosis, cell cycle, CSCs expression, and epithelial phenotype promotion. Taken all together, in this study we could demonstrate the extraordinary anticancer properties of NHC-based carbosilane dendrimers against androgen-resistant prostate cancer cells in vitro.
Collapse
Affiliation(s)
- Oscar Barrios
- University of Alcalá, Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. Del Río" (IQAR), Madrid, 28871, Spain
| | - Belén G Sánchez
- University of Alcalá, Biochemistry and Molecular Biology Unit. Department of Systems Biology and Research Institute in Chemistry "Andrés M. Del Río" (IQAR), Madrid, 28871, Spain
| | - Tamara Rodríguez-Prieto
- University of Alcalá, Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. Del Río" (IQAR), Madrid, 28871, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, 28029, Spain.,Ramón y Cajal Health Research Institute (IRYCIS), IRYCIS, Madrid, 28034, Spain
| | - Jesús Cano
- University of Alcalá, Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. Del Río" (IQAR), Madrid, 28871, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, 28029, Spain.,Ramón y Cajal Health Research Institute (IRYCIS), IRYCIS, Madrid, 28034, Spain
| | - Alicia Bort
- University of Alcalá, Biochemistry and Molecular Biology Unit. Department of Systems Biology and Research Institute in Chemistry "Andrés M. Del Río" (IQAR), Madrid, 28871, Spain.,Yale University School of Medicine, Vascular Biology and Therapeutics Program, New Haven, Connecticut 06520, United States
| | - Rafael Gómez
- University of Alcalá, Department of Organic and Inorganic Chemistry, and Research Institute in Chemistry "Andrés M. Del Río" (IQAR), Madrid, 28871, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, 28029, Spain.,Ramón y Cajal Health Research Institute (IRYCIS), IRYCIS, Madrid, 28034, Spain
| | - Inés Díaz-Laviada
- University of Alcalá, Biochemistry and Molecular Biology Unit. Department of Systems Biology and Research Institute in Chemistry "Andrés M. Del Río" (IQAR), Madrid, 28871, Spain
| |
Collapse
|
26
|
Chhetri D, Vengadassalapathy S, Venkadassalapathy S, Balachandran V, Umapathy VR, Veeraraghavan VP, Jayaraman S, Patil S, Iyaswamy A, Palaniyandi K, Gnanasampanthapandian D. Pleiotropic effects of DCLK1 in cancer and cancer stem cells. Front Mol Biosci 2022; 9:965730. [PMID: 36250024 PMCID: PMC9560780 DOI: 10.3389/fmolb.2022.965730] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 08/12/2022] [Indexed: 12/02/2022] Open
Abstract
Doublecortin-like kinase 1 (DCLK1), a protein molecule, has been identified as a tumor stem cell marker in the cancer cells of gastrointestinal, pancreas, and human colon. DCLK1 expression in cancers, such as breast carcinoma, lung carcinoma, hepatic cell carcinoma, tuft cells, and human cholangiocarcinoma, has shown a way to target the DCLK1 gene and downregulate its expression. Several studies have discussed the inhibition of tumor cell proliferation along with neoplastic cell arrest when the DCLK1 gene, which is expressed in both cancer and normal cells, was targeted successfully. In addition, previous studies have shown that DCLK1 plays a vital role in various cancer metastases. The correlation of DCLK1 with numerous stem cell receptors, signaling pathways, and genes suggests its direct or an indirect role in promoting tumorigenesis. Moreover, the impact of DCLK1 was found to be related to the functioning of an oncogene. The downregulation of DCLK1 expression by using targeted strategies, such as embracing the use of siRNA, miRNA, CRISPR/Cas9 technology, nanomolecules, specific monoclonal antibodies, and silencing the pathways regulated by DCLK1, has shown promising results in both in vitro and in vivo studies on gastrointestinal (GI) cancers. In this review, we will discuss about the present understanding of DCLK1 and its role in the progression of GI cancer and metastasis.
Collapse
Affiliation(s)
- Dibyashree Chhetri
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
| | - Srinivasan Vengadassalapathy
- Department of Pharmacology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | | | - Varadharaju Balachandran
- Department of Physiology, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Vidhya Rekha Umapathy
- Department of Public Health Dentistry, Sree Balaji Dental College and Hospital, Chennai, India
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Selvaraj Jayaraman
- Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Shankargouda Patil
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT, United States
| | - Ashok Iyaswamy
- Centre for Parkinsons Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Kanagaraj Palaniyandi
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
- *Correspondence: Kanagaraj Palaniyandi, ; Dhanavathy Gnanasampanthapandian,
| | - Dhanavathy Gnanasampanthapandian
- Cancer Science Laboratory, Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Chennai, India
- *Correspondence: Kanagaraj Palaniyandi, ; Dhanavathy Gnanasampanthapandian,
| |
Collapse
|
27
|
Park S, Park JM, Park M, Ko D, Kim S, Seo J, Nam KD, Jung E, Farrand L, Kim YJ, Kim JY, Seo JH. β-Escin overcomes trastuzumab resistance in HER2-positive breast cancer by targeting cancer stem-like features. Cancer Cell Int 2022; 22:289. [PMID: 36127671 PMCID: PMC9490928 DOI: 10.1186/s12935-022-02713-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 09/09/2022] [Indexed: 11/15/2022] Open
Abstract
Background The emergence of de novo or intrinsic trastuzumab resistance is exceedingly high in breast cancer that is HER2 positive and correlates with an abundant cancer stem cell (CSC)-like population. We sought to examine the capacity of β-escin, an anti-inflammatory drug, to address trastuzumab resistance in HER2-positive breast cancer cells. Methods The effect of β-escin on trastuzumab-resistant and -sensitive cell lines in vitro was evaluated for apoptosis, expression of HER2 family members, and impact on CSC-like properties. An in vivo model of trastuzumab-resistant JIMT-1 was used to examine the efficacy and toxicity of β-escin. Results β-escin induced mitochondrial-mediated apoptosis accompanied by reactive oxygen species (ROS) production and increased active p18Bax fragmentation, leading to caspase-3/-7 activation. Attenuation of CSC-related features by β-escin challenge was accompanied by marked reductions in CD44high/CD24low stem-like cells and aldehyde dehydrogenase 1 (ALDH1) activity as well as hindrance of mammosphere formation. β-escin administration also significantly retarded tumor growth and angiogenesis in a trastuzumab-resistant JIMT-1 xenograft model via downregulation of CSC-associated markers and intracellular domain HER2. Importantly, β-escin selectively inhibited malignant cells and was less toxic to normal mammary cells, and no toxic effects were found in liver and kidney function in animals. Conclusions Taken together, our findings highlight β-escin as a promising candidate for the treatment of trastuzumab-resistant HER2-positive breast cancers. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02713-9.
Collapse
Affiliation(s)
- Soeun Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Jung Min Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Minsu Park
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Dongmi Ko
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Seongjae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Juyeon Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Kee Dal Nam
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Eunsun Jung
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea.,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea
| | - Lee Farrand
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, 5000, Australia
| | - Yoon-Jae Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea. .,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea. .,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea.
| | - Ji Young Kim
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea. .,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea.
| | - Jae Hong Seo
- Division of Medical Oncology, Department of Internal Medicine, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea. .,Brain Korea 21 Program for Biomedical Science, Korea University College of Medicine, Korea University, Seoul, 02841, Republic of Korea. .,Department of Biomedical Research Center, Korea University Guro Hospital, Korea University, 97 Gurodong-gil, Guro-gu, Seoul, 08308, Republic of Korea.
| |
Collapse
|
28
|
Gomez RL, Woods LM, Ramachandran R, Abou Tayoun AN, Philpott A, Ali FR. Super-enhancer associated core regulatory circuits mediate susceptibility to retinoic acid in neuroblastoma cells. Front Cell Dev Biol 2022; 10:943924. [PMID: 36147741 PMCID: PMC9485839 DOI: 10.3389/fcell.2022.943924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 08/08/2022] [Indexed: 11/25/2022] Open
Abstract
Neuroblastoma is a pediatric tumour that accounts for more than 15% of cancer-related deaths in children. High-risk tumours are often difficult to treat, and patients' survival chances are less than 50%. Retinoic acid treatment is part of the maintenance therapy given to neuroblastoma patients; however, not all tumours differentiate in response to retinoic acid. Within neuroblastoma tumors, two phenotypically distinct cell types have been identified based on their super-enhancer landscape and transcriptional core regulatory circuitries: adrenergic (ADRN) and mesenchymal (MES). We hypothesized that the distinct super-enhancers in these different tumour cells mediate differential response to retinoic acid. To this end, three different neuroblastoma cell lines, ADRN (MYCN amplified and non-amplified) and MES cells, were treated with retinoic acid, and changes in the super-enhancer landscape upon treatment and after subsequent removal of retinoic acid was studied. Using ChIP-seq for the active histone mark H3K27ac, paired with RNA-seq, we compared the super-enhancer landscape in cells that undergo neuronal differentiation in response to retinoic acid versus those that fail to differentiate and identified unique super-enhancers associated with neuronal differentiation. Among the ADRN cells that respond to treatment, MYCN-amplified cells remain differentiated upon removal of retinoic acid, whereas MYCN non-amplified cells revert to an undifferentiated state, allowing for the identification of super-enhancers responsible for maintaining differentiation. This study identifies key super-enhancers that are crucial for retinoic acid-mediated differentiation.
Collapse
Affiliation(s)
- Roshna Lawrence Gomez
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Laura M. Woods
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Center, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Revathy Ramachandran
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Ahmad N. Abou Tayoun
- Center for Genomic Discovery, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Al Jalila Genomics Center, Al Jalila Children’s Hospital, Dubai, United Arab Emirates
| | - Anna Philpott
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Center, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Fahad R. Ali
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
- Center for Genomic Discovery, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| |
Collapse
|
29
|
Mokkapati S, Narayan VM, Manyam GC, Lim AH, Duplisea JJ, Kokorovic A, Miest TS, Mitra AP, Plote D, Anand SS, Metcalfe MJ, Dunner K, Johnson BA, Czerniak BA, Nieminen T, Heikura T, Yla-Herttuala S, Parker NR, Schluns KS, McConkey DJ, Dinney CP. Lentiviral interferon: A novel method for gene therapy in bladder cancer. Mol Ther Oncolytics 2022; 26:141-157. [PMID: 35847448 PMCID: PMC9251210 DOI: 10.1016/j.omto.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 06/07/2022] [Indexed: 11/29/2022] Open
Abstract
Interferon alpha (IFNα) gene therapy is emerging as a new treatment option for patients with non-muscle invasive bladder cancer (NMIBC). Adenoviral vectors expressing IFNα have shown clinical efficacy treating bacillus Calmette-Guerin (BCG)-unresponsive bladder cancer (BLCA). However, transient transgene expression and adenoviral immunogenicity may limit therapeutic activity. Lentiviral vectors can achieve stable transgene expression and are less immunogenic. In this study, we evaluated lentiviral vectors expressing murine IFNα (LV-IFNα) and demonstrate IFNα expression by transduced murine BLCA cell lines, bladder urothelium, and within the urine following intravesical instillation. Murine BLCA cell lines (MB49 and UPPL1541) were sensitive to IFN-mediated cell death after LV-IFNα, whereas BBN975 was inherently resistant. Upregulation of interleukin-6 (IL-6) predicted sensitivity to IFN-mediated cell death mediated by caspase signaling, which when inhibited abrogated IFN-mediated cell killing. Intravesical therapy with LV-IFNα/Syn3 in a syngeneic BLCA model significantly improved survival, and molecular analysis of treated tumors revealed upregulation of apoptotic and immune-cell-mediated death pathways. In particular, biomarker discovery analysis identified three clinically actionable targets, PD-L1, epidermal growth factor receptor (EGFR), and ALDHA1A, in murine tumors treated with LV-IFNα/Syn3. Our findings warrant the comparison of adenoviral and LV-IFNα and the study of novel combination strategies with IFNα gene therapy for the BLCA treatment.
Collapse
Affiliation(s)
- Sharada Mokkapati
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
- Corresponding author Sharada Mokkapati, PhD, University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA.
| | - Vikram M. Narayan
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - Ganiraju C. Manyam
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - Amy H. Lim
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - Jonathan J. Duplisea
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - Andrea Kokorovic
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - Tanner S. Miest
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - Anirban P. Mitra
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - Devin Plote
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - Selvalakshmi Selvaraj Anand
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - Michael J. Metcalfe
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - Kenneth Dunner
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - Burles A. Johnson
- James Buchanan Brady Urological Institute, John Hopkins Greenberg Bladder Cancer Institute, John Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Bogdan A. Czerniak
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - Tiina Nieminen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tommi Heikura
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - Seppo Yla-Herttuala
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | | | - Kimberley S. Schluns
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
| | - David J. McConkey
- James Buchanan Brady Urological Institute, John Hopkins Greenberg Bladder Cancer Institute, John Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Colin P. Dinney
- University of Texas MD Anderson Cancer Center, Smith Research Building, 7777 Knight Road, Houston, TX 77584, USA
- Corresponding author Colin P. Dinney, MD, University of Texas MD Anderson Cancer Center, CPB7.3279, 1515 Holcombe Blvd., Houston, TX 77030, USA.
| |
Collapse
|
30
|
Curcumin-based-fluorescent probes targeting ALDH1A3 as a promising tool for glioblastoma precision surgery and early diagnosis. Commun Biol 2022; 5:895. [PMID: 36050388 PMCID: PMC9437101 DOI: 10.1038/s42003-022-03834-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive primary brain tumour for which both effective treatments and efficient tools for an early-stage diagnosis are lacking. Herein, we present curcumin-based fluorescent probes that are able to bind to aldehyde dehydrogenase 1A3 (ALDH1A3), an enzyme overexpressed in glioma stem cells (GSCs) and associated with stemness and invasiveness of GBM. Two compounds are selective versus ALDH1A3, without showing any appreciable interaction with other ALDH1A isoenzymes. Indeed, their fluorescent signal is detectable only in our positive controls in vitro and absent in cells that lack ALDH1A3. Remarkably, in vivo, our Probe selectively accumulate in glioblastoma cells, allowing the identification of the growing tumour mass. The significant specificity of our compounds is the necessary premise for their further development into glioblastoma cells detecting probes to be possibly used during neurosurgical operations. Curcumin-based fluorescent probes are presented that are able to selectively bind to aldehyde dehydrogenase 1A3 (ALDH1A3), an enzyme overexpressed in glioma stem cells and specifically identify glioblastoma cells in vitro and in vivo.
Collapse
|
31
|
Isothiocyanates (ITCs) 1-(Isothiocyanatomethyl)-4-phenylbenzene and 1-Isothiocyanato-3,5-bis(trifluoromethyl)benzene—Aldehyde Dehydrogenase (ALDH) Inhibitors, Decreases Cisplatin Tolerance and Migratory Ability of NSCLC. Int J Mol Sci 2022; 23:ijms23158644. [PMID: 35955773 PMCID: PMC9369118 DOI: 10.3390/ijms23158644] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 01/27/2023] Open
Abstract
One of the main treatment modalities for non-small-cell lung cancer (NSCLC) is cisplatin-based chemotherapy. However, the acquisition of cisplatin resistance remains a major problem. Existing chemotherapy regimens are often ineffective against cancer cells expressing aldehyde dehydrogenase (ALDH). As such, there is an urgent need for therapies targeting ALDH-positive cancer cells. The present study compares the anticancer properties of 36 structurally diverse isothiocyanates (ITCs) against NSCLC cells with the ALDH inhibitor disulfiram (DSF). Their potential affinity to ALDH isoforms and ABC proteins was assessed using AutoDockTools, allowing for selection of three compounds presenting the strongest affinity to all tested proteins. The selected ITCs had no impact on NSCLC cell viability (at tested concentrations), but significantly decreased the cisplatin tolerance of cisplatin-resistant variant of A549 (A549CisR) and advanced (stage 4) NSCLC cell line H1581. Furthermore, long-term supplementation with ITC 1-(isothiocyanatomethyl)-4-phenylbenzene reverses the EMT phenotype and migratory potential of A549CisR to the level presented by parental A549 cells, increasing E-Cadherin expression, followed by decreased expression of ABCC1 and ALDH3A1. Our data indicates that the ALDH inhibitors DSF and ITCs are potential adjuvants of cisplatin chemotherapy.
Collapse
|
32
|
Regulation of the Cancer Stem Phenotype by Long Non-Coding RNAs. Cells 2022; 11:cells11152352. [PMID: 35954194 PMCID: PMC9367355 DOI: 10.3390/cells11152352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/21/2022] [Accepted: 07/24/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer stem cells are a cell population within malignant tumors that are characterized by the ability to self-renew, the presence of specific molecules that define their identity, the ability to form malignant tumors in vivo, resistance to drugs, and the ability to invade and migrate to other regions of the body. These characteristics are regulated by various molecules, such as lncRNAs, which are transcripts that generally do not code for proteins but regulate multiple biological processes through various mechanisms of action. LncRNAs, such as HOTAIR, H19, LncTCF7, LUCAT1, MALAT1, LINC00511, and FMR1-AS1, have been described as key regulators of stemness in cancer, allowing cancer cells to acquire this phenotype. It has been proposed that cancer stem cells are clinically responsible for the high recurrence rates after treatment and the high frequency of metastasis in malignant tumors, so understanding the mechanisms that regulate the stem phenotype could have an impact on the improvement of cancer treatments.
Collapse
|
33
|
Dhanyamraju PK, Schell TD, Amin S, Robertson GP. Drug-Tolerant Persister Cells in Cancer Therapy Resistance. Cancer Res 2022; 82:2503-2514. [PMID: 35584245 PMCID: PMC9296591 DOI: 10.1158/0008-5472.can-21-3844] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 03/15/2022] [Accepted: 05/09/2022] [Indexed: 01/21/2023]
Abstract
One of the current stumbling blocks in our fight against cancer is the development of acquired resistance to therapy, which is attributable to approximately 90% of cancer-related deaths. Undercutting this process during treatment could significantly improve cancer management. In many cases, drug resistance is mediated by a drug-tolerant persister (DTP) cell subpopulation present in tumors, often referred to as persister cells. This review provides a summary of currently known persister cell subpopulations and approaches to target them. A specific DTP cell subpopulation with elevated levels of aldehyde dehydrogenase (ALDH) activity has stem cell-like characteristics and a high level of plasticity, enabling them to switch rapidly between high and low ALDH activity. Further studies are required to fully elucidate the functions of ALDH-high DTP cells, how they withstand drug concentrations that kill other cells, and how they rapidly adapt under levels of high cellular stress and eventually lead to more aggressive, recurrent, and drug-resistant cancer. Furthermore, this review addresses the processes used by the ALDH-high persister cell subpopulation to enable cancer progression, the ALDH isoforms important in these processes, interactions of ALDH-high DTPs with the tumor microenvironment, and approaches to therapeutically modulate this subpopulation in order to more effectively manage cancer.
Collapse
Affiliation(s)
- Pavan Kumar Dhanyamraju
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Todd D Schell
- Departments of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Shantu Amin
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - Gavin P Robertson
- Department of Pharmacology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Pathology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Dermatology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Department of Surgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- The Penn State Melanoma and Skin Cancer Center, The Pennsylvania State University College of Medicine, Hershey, PA 17033
- Penn State Melanoma Therapeutics Program, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| |
Collapse
|
34
|
Lu HJ, Chuang CY, Chen MK, Su CW, Yang WE, Yeh CM, Lai KM, Tang CH, Lin CW, Yang SF. The impact of ALDH7A1 variants in oral cancer development and prognosis. Aging (Albany NY) 2022; 14:4556-4571. [PMID: 35613852 PMCID: PMC9186774 DOI: 10.18632/aging.204099] [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: 11/04/2021] [Accepted: 05/19/2022] [Indexed: 11/25/2022]
Abstract
The gene encoding aldehyde dehydrogenase 7 family member A1 (ALDH7A1) has been associated with the development and prognosis in multiple cancers; however, the role of ALDH7A1 polymorphisms in oral cancer remains unknown. For this purpose, the influences of ALDH7A1 rs13182402 and rs12659017 on oral cancer development and prognosis were analyzed. Our resulted showed that ALDH7A1 rs13182402 genotype had less pathologic nodal metastasis among betel quid chewer. ALDH7A1 rs13182402 also corresponded to higher expressions in upper aerodigestive mucosa, whole blood, the musculoskeletal system and oral cancer tissues than did the ALDH7A1 wild type. Furthermore, ALDH7A1 overexpression in oral cancer cells increased in vitro migration, whereas its silencing reduced cell migration. Conversely, ALDH7A1 expression in tumor tissues and in patients with advanced disease was lower than that in normal tissues and in patients with early-stage disease. When the patients were classified into ALDH7A1-high and -low-expression groups, the high-ALDH7A1 group had superior outcomes in progression-free survival than the low-ALDH7A1 group (5-year survival of 58.7% vs. 48.0%, P = 0.048) did. In conclusion, patients with high ALDH7A1 expression might, however, have more favorable prognoses than those with low ALDH7A1 expression have.
Collapse
Affiliation(s)
- Hsueh-Ju Lu
- Division of Hematology and Oncology, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Yi Chuang
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Otolaryngology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Mu-Kuan Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua, Taiwan.,Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Chun-Wen Su
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Wei-En Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chia-Ming Yeh
- Oral Cancer Research Center, Changhua Christian Hospital, Changhua, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Kuan-Ming Lai
- Division of Hematology and Oncology, Department of Medicine, Changhua Christian Hospital, Changhua, Taiwan
| | - Chih-Hsin Tang
- School of Medicine, China Medical University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Chiao-Wen Lin
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan.,Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
35
|
Kao Y, Huang LC, Hsu SY, Huang SM, Hueng DY. The Effect of Disulfiram and Copper on Cellular Viability, ER Stress and ALDH Expression of Human Meningioma Cells. Biomedicines 2022; 10:887. [PMID: 35453636 PMCID: PMC9025959 DOI: 10.3390/biomedicines10040887] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/27/2022] [Accepted: 04/06/2022] [Indexed: 11/17/2022] Open
Abstract
(1) Background: Meningiomas are the most common intracranial tumors in adults; currently there is no effective chemotherapy for malignant meningiomas. The effect of disulfiram (DSF)/Copper (Cu) on meningiomas remains unclear; (2) Methods: The impact of DSF/Cu on cell viability of meningioma adhesion cells (MgACs) and sphere cells (MgSCs) was assessed via MTS assay. The effects of DSF/Cu on intracellular Cu levels, cell senescence, and apoptosis were analyzed using CopperGreen, C12FDG, and Annexin V assays. Intracellular ALDH isoform expression and canonical pathway expression after DSF/Cu treatment were analyzed using mRNA microarray and Ingenuity Pathway Analysis, with further verification through qRT-PCR and immunoblotting; (3) Results: The viability of MgACs and MgSCs were inhibited by DSF/Cu. DSF/Cu increased intracellular Cu levels and cellular senescence. DSF/Cu also induced ER stress in MgACs and activated the PERK/eIF2 pathway for further adaptive response, apoptosis, and autophagy. Finally, DSF/Cu inhibited the expression of different ALDH isoforms in MgACs and MgSCs; (4) Conclusions: DSF/Cu exerts cytotoxic effects against both meningioma cells and stem-like cells and has treatment potential for meningioma.
Collapse
Affiliation(s)
- Ying Kao
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan;
- Division of Neurosurgery, Department of Surgery, Taipei City Hospital Zhongxing Branch, Taipei 10341, Taiwan
| | - Li-Chun Huang
- Department of Biochemistry, National Defense Medical Center, Taipei 11490, Taiwan; (L.-C.H.); (S.-M.H.)
| | - Shao-Yuan Hsu
- Division of Neurosurgery, Department of Surgery, Taipei City Hospital Renai Branch, Taipei 106243, Taiwan;
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei 11490, Taiwan; (L.-C.H.); (S.-M.H.)
| | - Dueng-Yuan Hueng
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 11490, Taiwan;
- Department of Biochemistry, National Defense Medical Center, Taipei 11490, Taiwan; (L.-C.H.); (S.-M.H.)
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
| |
Collapse
|
36
|
Modulation of Notch Signaling Pathway by Bioactive Dietary Agents. Int J Mol Sci 2022; 23:ijms23073532. [PMID: 35408894 PMCID: PMC8998406 DOI: 10.3390/ijms23073532] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/19/2022] [Accepted: 03/22/2022] [Indexed: 12/15/2022] Open
Abstract
Notch signaling is often aberrantly activated in solid and hematological cancers and regulates cell fate decisions and the maintenance of cancer stem cells. In addition, increased expression of Notch pathway components is clinically associated with poorer prognosis in several types of cancer. Targeting Notch may have chemopreventive and anti-cancer effects, leading to reduced disease incidence and improved survival. While therapeutic agents are currently in development to achieve this goal, several researchers have turned their attention to dietary and natural agents for targeting Notch signaling. Given their natural abundance from food sources, the use of diet-derived agents to target Notch signaling offers the potential advantage of low toxicity to normal tissue. In this review, we discuss several dietary agents including curcumin, EGCG, resveratrol, and isothiocyanates, which modulate Notch pathway components in a context-dependent manner. Dietary agents modulate Notch signaling in several types of cancer and concurrently decrease in vitro cell viability and in vivo tumor growth, suggesting a potential role for their clinical use to target Notch pathway components, either alone or in combination with current therapeutic agents.
Collapse
|
37
|
Hadjis AD, Nunes NS, Khan SM, Fletcher RE, Pohl ADP, Venzon DJ, Eckhaus MA, Kanakry CG. Post-Transplantation Cyclophosphamide Uniquely Restrains Alloreactive CD4 + T-Cell Proliferation and Differentiation After Murine MHC-Haploidentical Hematopoietic Cell Transplantation. Front Immunol 2022; 13:796349. [PMID: 35242129 PMCID: PMC8886236 DOI: 10.3389/fimmu.2022.796349] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/06/2022] [Indexed: 12/25/2022] Open
Abstract
Post-transplantation cyclophosphamide (PTCy) reduces the incidence and severity of graft-versus-host disease (GVHD), thereby improving the safety and accessibility of allogeneic hematopoietic cell transplantation (HCT). We have shown that PTCy works by inducing functional impairment and suppression of alloreactive T cells. We also have identified that reduced proliferation of alloreactive CD4+ T cells at day +7 and preferential recovery of CD4+CD25+Foxp3+ regulatory T cells (Tregs) at day +21 are potential biomarkers associated with optimal PTCy dosing and timing in our B6C3F1→B6D2F1 MHC-haploidentical murine HCT model. To understand whether the effects of PTCy are unique and also to understand better the biology of GVHD prevention by PTCy, here we tested the relative impact of cyclophosphamide compared with five other optimally dosed chemotherapeutics (methotrexate, bendamustine, paclitaxel, vincristine, and cytarabine) that vary in mechanisms of action and drug resistance. Only cyclophosphamide, methotrexate, and cytarabine were effective in preventing fatal GVHD, but cyclophosphamide was superior in ameliorating both clinical and histopathological GVHD. Flow cytometric analyses of blood and spleens revealed that these three chemotherapeutics were distinct in constraining conventional T-cell numerical recovery and facilitating preferential Treg recovery at day +21. However, cyclophosphamide was unique in consistently reducing proliferation and expression of the activation marker CD25 by alloreactive CD4+Foxp3- conventional T cells at day +7. Furthermore, cyclophosphamide restrained the differentiation of alloreactive CD4+Foxp3- conventional T cells at both days +7 and +21, whereas methotrexate and cytarabine only restrained differentiation at day +7. No chemotherapeutic selectively eliminated alloreactive T cells. These data suggest that constrained alloreactive CD4+Foxp3- conventional T-cell numerical recovery and associated preferential CD4+CD25+Foxp3+ Treg reconstitution at day +21 may be potential biomarkers of effective GVHD prevention. Additionally, these results reveal that PTCy uniquely restrains alloreactive CD4+Foxp3- conventional T-cell proliferation and differentiation, which may explain the superior effects of PTCy in preventing GVHD. Further study is needed to determine whether these findings also hold true in clinical HCT.
Collapse
Affiliation(s)
- Ashley D. Hadjis
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Natalia S. Nunes
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Shanzay M. Khan
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Rochelle E. Fletcher
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Alessandra de Paula Pohl
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - David J. Venzon
- Biostatistics and Data Management Section, Office of the Clinical Director, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Michael A. Eckhaus
- Division of Veterinary Resources, Office of Research Services, National Institutes of Health, Bethesda, MD, United States
| | - Christopher G. Kanakry
- Experimental Transplantation and Immunotherapy Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
38
|
Won M, Kim JH, Ji MS, Kim JS. ROS activated prodrug for ALDH overexpressed cancer stem cells. Chem Commun (Camb) 2021; 58:72-75. [PMID: 34874378 DOI: 10.1039/d1cc05573a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aldehyde dehydrogenase (ALDH), a cancer stem cell biomarker, is related to drug resistance. Co-treatment of anti-cancer drug (CPT) and ALDH inhibitor (DEAB) can overcome the drug resistance of cancer stem cells (CSCs) and finally cure cancers without relapse. We herein introduce a prodrug (DE-CPT) - consisting of 1,3-oxathiolane as an ROS responsive scaffold, and an aldehyde protecting group of DEAB - to deliver the CPT and DEAB upon reaction with ROS. From tests of the sphere-forming ability and CSC marker subpopulation, we found that DE-CPT efficiently decreases the CSCs population and kills the cancer cells.
Collapse
Affiliation(s)
- Miae Won
- Department of Chemistry, Korea University, Seoul 02841, Korea.
| | - Ji Hyeon Kim
- Department of Chemistry, Korea University, Seoul 02841, Korea.
| | - Myung Sun Ji
- Department of Chemistry, Korea University, Seoul 02841, Korea.
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul 02841, Korea.
| |
Collapse
|
39
|
Yang H, Hou H, Zhao H, Yu T, Hu Y, Hu Y, Guo J. HK2 Is a Crucial Downstream Regulator of miR-148a for the Maintenance of Sphere-Forming Property and Cisplatin Resistance in Cervical Cancer Cells. Front Oncol 2021; 11:794015. [PMID: 34858863 PMCID: PMC8631922 DOI: 10.3389/fonc.2021.794015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 10/25/2021] [Indexed: 01/10/2023] Open
Abstract
The acquisition of cancer stem-like properties is believed to be responsible for cancer metastasis and therapeutic resistance in cervical cancer (CC). CC tissues display a high expression level of hexokinase 2 (HK2), which is critical for the proliferation and migration of CC cells. However, little is known about the functional role of HK2 in the maintenance of cancer stem cell-like ability and cisplatin resistance of CC cells. Here, we showed that the expression of HK2 is significantly elevated in CC tissues, and high HK2 expression correlates with poor prognosis. HK2 overexpression (or knockdown) can promote (or inhibit) the sphere-forming ability and cisplatin resistance in CC cells. In addition, HK2-overexpressing CC cells show enhanced expression of cancer stem cell-associated genes (including SOX2 and OCT4) and drug resistance-related gene MDR1. The expression of HK2 is mediated by miR-145, miR-148a, and miR-497 in CC cells. Overexpression of miR-148a is sufficient to reduce sphere formation and cisplatin resistance in CC cells. Our results elucidate a novel mechanism through which miR-148a regulates CC stem cell-like properties and chemoresistance by interfering with the oncogene HK2, providing the first evidence that dysregulation of the miR-148a/HK2 signaling plays a critical role in the maintenance of sphere formation and cisplatin resistance of CC cells. Our findings may guide future studies on therapeutic strategies that reverse cisplatin resistance by targeting this pathway.
Collapse
Affiliation(s)
- Hao Yang
- Department of Radiation Oncology, Inner Mongolia Cancer Hospital and Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hui Hou
- Department of Pediatric Hematology and Oncology, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Haiping Zhao
- Department of Abdominal Tumor Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Tianwei Yu
- Department of Transfusion Medicine, Inner Mongolia Cancer Hospital and Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yuchong Hu
- Department of Gynaecology, Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Yue Hu
- Department of Radiation Oncology, Inner Mongolia Cancer Hospital and Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Junmei Guo
- Department of Radiation Oncology, Inner Mongolia Cancer Hospital and Affiliated People's Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
40
|
Colombo G, Gelardi ELM, Balestrero FC, Moro M, Travelli C, Genazzani AA. Insight Into Nicotinamide Adenine Dinucleotide Homeostasis as a Targetable Metabolic Pathway in Colorectal Cancer. Front Pharmacol 2021; 12:758320. [PMID: 34880756 PMCID: PMC8645963 DOI: 10.3389/fphar.2021.758320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/02/2021] [Indexed: 11/13/2022] Open
Abstract
Tumour cells modify their cellular metabolism with the aim to sustain uncontrolled proliferation. Cancer cells necessitate adequate amounts of NAD and NADPH to support several enzymes that are usually overexpressed and/or overactivated. Nicotinamide adenine dinucleotide (NAD) is an essential cofactor and substrate of several NAD-consuming enzymes, such as PARPs and sirtuins, while NADPH is important in the regulation of the redox status in cells. The present review explores the rationale for targeting the key enzymes that maintain the cellular NAD/NADPH pool in colorectal cancer and the enzymes that consume or use NADP(H).
Collapse
Affiliation(s)
- Giorgia Colombo
- Department of Pharmaceutical Sciences, Università Del Piemonte Orientale, Novara, Italy
| | | | | | - Marianna Moro
- Department of Pharmaceutical Sciences, Università Del Piemonte Orientale, Novara, Italy
| | - Cristina Travelli
- Department of Drug Sciences, Università Degli Studi di Pavia, Pavia, Italy
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences, Università Del Piemonte Orientale, Novara, Italy
| |
Collapse
|
41
|
Hsueh WT, Chen SH, Chien CH, Chou SW, Chi PI, Chu JM, Chang KY. SOD2 Enhancement by Long-Term Inhibition of the PI3K Pathway Confers Multi-Drug Resistance and Enhanced Tumor-Initiating Features in Head and Neck Cancer. Int J Mol Sci 2021; 22:ijms222011260. [PMID: 34681918 PMCID: PMC8537886 DOI: 10.3390/ijms222011260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/15/2021] [Accepted: 10/16/2021] [Indexed: 11/16/2022] Open
Abstract
The phosphoinositide-3-kinase (PI3K) pathway has widely been considered as a potential therapeutic target for head and neck cancer (HNC); however, the application of PI3K inhibitors is often overshadowed by the induction of drug resistance with unknown mechanisms. In this study, PII3K inhibitor resistant cancer cells were developed by prolonged culturing of cell lines with BEZ235, a dual PI3K and mammalian target of rapamycin (mTOR) inhibitor. The drug resistant HNC cells showed higher IC50 of the proliferation to inhibitors specifically targeting PI3K and/or mTOR, as compared to their parental cells. These cells also showed profound resistance to drugs of other classes. Molecular analysis revealed persistent activation of phosphorylated AKT at threonine 308 in the drug resistant cells and increased expression of markers for tumor-initiating cells. Interestingly, increased intra-cellular ROS levels were observed in the drug resistant cells. Among anti-oxidant molecules, the expression of SOD2 was increased and was associated with the ALDH-positive tumor-initiating cell features. Co-incubation of SOD inhibitors and BEZ235 decreased the stemness feature of the cells in vitro, as shown by results of the spheroid formation assay. In conclusion, dysregulation of SOD2 might contribute to the profound resistance to PI3K inhibitors and the other drugs in HNC cells.
Collapse
Affiliation(s)
- Wei-Ting Hsueh
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70456, Taiwan; (W.-T.H.); (S.-H.C.)
| | - Shang-Hung Chen
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70456, Taiwan; (W.-T.H.); (S.-H.C.)
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan; (C.-H.C.); (S.-W.C.); (P.-I.C.); (J.-M.C.)
| | - Chia-Hung Chien
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan; (C.-H.C.); (S.-W.C.); (P.-I.C.); (J.-M.C.)
- School of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| | - Shao-Wen Chou
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan; (C.-H.C.); (S.-W.C.); (P.-I.C.); (J.-M.C.)
| | - Pei-I Chi
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan; (C.-H.C.); (S.-W.C.); (P.-I.C.); (J.-M.C.)
| | - Jui-Mei Chu
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan; (C.-H.C.); (S.-W.C.); (P.-I.C.); (J.-M.C.)
| | - Kwang-Yu Chang
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 70456, Taiwan; (W.-T.H.); (S.-H.C.)
- National Institute of Cancer Research, National Health Research Institutes, Tainan 70456, Taiwan; (C.-H.C.); (S.-W.C.); (P.-I.C.); (J.-M.C.)
- Correspondence: ; Tel.: +886-6-208-3422
| |
Collapse
|
42
|
Liu CC, Wu CL, Lin MX, Sze CI, Gean PW. Disulfiram Sensitizes a Therapeutic-Resistant Glioblastoma to the TGF-β Receptor Inhibitor. Int J Mol Sci 2021; 22:ijms221910496. [PMID: 34638842 PMCID: PMC8508702 DOI: 10.3390/ijms221910496] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/16/2021] [Accepted: 09/24/2021] [Indexed: 02/08/2023] Open
Abstract
Despite neurosurgery following radiation and chemotherapy, residual glioblastoma (GBM) cells develop therapeutic resistance (TR) leading to recurrence. The GBM heterogeneity confers TR. Therefore, an effective strategy must target cancer stem cells (CSCs) and other malignant cancer cells. TGF-β and mesenchymal transition are the indicators for poor prognoses. The activity of aldehyde dehydrogenases (ALDHs) is a functional CSC marker. However, the interplay between TGF-β and ALDHs remains unclear. We developed radiation-resistant and radiation-temozolomide-resistant GBM models to investigate the underlying mechanisms conferring TR. Galunisertib is a drug targeting TGF-β receptors. Disulfiram (DSF) is an anti-alcoholism drug which functions by inhibiting ALDHs. The anti-tumor effects of combining DSF and Galunisertib were evaluated by in vitro cell grow, wound healing, Transwell assays, and in vivo orthotopic GBM model. Mesenchymal-like phenotype was facilitated by TGF-β in TR GBM. Additionally, TR activated ALDHs. DSF inhibited TR-induced cell migration and tumor sphere formation. However, DSF did not affect the tumor growth in vivo. Spectacularly, DSF sensitized TR GBM to Galunisertib both in vitro and in vivo. ALDH activity positively correlated with TGF-β-induced mesenchymal properties in TR GBM. CSCs and mesenchymal-like GBM cells targeted together by combining DSF and Galunisertib may be a good therapeutic strategy for recurrent GBM patients.
Collapse
Affiliation(s)
- Chan-Chuan Liu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan;
| | - Cheng-Lin Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng-Kung University, Tainan 701, Taiwan;
- Department of Pathology, College of Medicine, National Cheng Kung University Hospital, National Cheng-Kung University, Tainan 701, Taiwan
| | - Meng-Xuan Lin
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan;
| | - Chun-I Sze
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan;
- Department of Pathology, College of Medicine, National Cheng Kung University Hospital, National Cheng-Kung University, Tainan 701, Taiwan
- Department of Cell Biology and Anatomy, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan;
- Correspondence: (C.-I.S.); (P.-W.G.)
| | - Po-Wu Gean
- Institute of Basic Medical Sciences, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan;
- Department of Pharmacology, College of Medicine, National Cheng-Kung University, Tainan 701, Taiwan
- Department of Biotechnology and Bioindustry Sciences, National Cheng-Kung University, Tainan 701, Taiwan
- Correspondence: (C.-I.S.); (P.-W.G.)
| |
Collapse
|
43
|
Poturnajova M, Kozovska Z, Matuskova M. Aldehyde dehydrogenase 1A1 and 1A3 isoforms - mechanism of activation and regulation in cancer. Cell Signal 2021; 87:110120. [PMID: 34428540 PMCID: PMC8505796 DOI: 10.1016/j.cellsig.2021.110120] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 12/15/2022]
Abstract
In some types of human cancer, aldehyde dehydrogenases represent stemness markers and their expression is associated with advanced disease stages and poor prognosis. Although several biological functions are mediated by their product Retinoid acid, the molecular mechanism is tissue-dependent and only partially understood. In this review, we summarize the current knowledge about the role of ALDH in solid tumours, especially ALDH1A1 and ALDH1A3 isoforms, regarding the molecular mechanism of their transcription and regulation, and their crosstalk with main molecular pathways resulting in the excessive proliferation, chemoresistance, stem cells properties and invasiveness. The recent knowledge of the regulatory effect of lnRNA on ALDH1A1 and ALDH1A3 is discussed too. Aldehyde dehydrogenases are important stem cell markers in many human cancer types. ALDH1A1 or ALDH1A3 activation participates in tumour progression, chemoresistance, stem-cell properties and invasiveness. ALDH1A1 interacts with oncogenic pathways Notch, NRF, CXCR4, Polycomb, MDR, and HOX.
Collapse
Affiliation(s)
- M Poturnajova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia.
| | - Z Kozovska
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia
| | - M Matuskova
- Department of Molecular Oncology, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, Dubravska cesta 9, 84505 Bratislava, Slovakia
| |
Collapse
|
44
|
Liu PF, Shu CW, Lee CH, Sie HC, Liou HH, Cheng JT, Ger LP, Chen CL, Chen CC, Chen CF. Clinical Significance and the Role of Guanylate-Binding Protein 5 in Oral Squamous Cell Carcinoma. Cancers (Basel) 2021; 13:cancers13164043. [PMID: 34439200 PMCID: PMC8394330 DOI: 10.3390/cancers13164043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/03/2021] [Accepted: 08/08/2021] [Indexed: 12/12/2022] Open
Abstract
Guanylate binding protein 5 (GBP5) is the interferon (IFN)-inducible subfamily of guanosine triphosphatases (GTPases) and is involved in pathogen defense. However, the role played by GBP5 in cancer development, especially in oral squamous cell carcinoma (OSCC), is still unknown. Herein, next-generation sequencing analysis showed that the gene expression levels of GBP5 were significantly higher in OSCC tissues compared with those found in corresponding tumor adjacent normal tissues (CTAN) from two pairs of OSCC patients. Higher gene expression levels of GBP5 were also found in tumor tissues of 23 buccal mucosal squamous cell carcinoma (BMSCC)/14 tongue squamous cell carcinoma (TSCC) patients and 30 oral cancer patients from The Cancer Genome Atlas (TCGA) database compared with those in CTAN tissues. Immunohistochemical results showed that protein expression levels of GBP5 were also higher in the tumor tissues of 353 OSCC patients including 117 BMSCC, 187 TSCC, and 49 lip squamous cell carcinoma patients. Moreover, TCGA database analysis indicated that high gene expression levels of GBP5 were associated with poor overall survival in oral cancer patients with moderate/poor cell differentiation, and associated with poor disease-free survival in oral cancer patients with moderate/poor cell differentiation and lymph node metastasis. Furthermore, GBP5-knockdowned cells exhibited decreased cell growth, arrest at G1 phase, and decreased invasion/migration. The gene expression of markers for epithelial-mesenchymal transition and cancer stemness was also reduced in GBP5-silenced oral cancer cells. Taken together, GBP5 might be a potential biomarker and therapeutic target for OSCC patients, especially for those with poor cell differentiation and lymph node metastasis.
Collapse
Affiliation(s)
- Pei-Feng Liu
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (P.-F.L.); or (C.-H.L.)
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Chih-Wen Shu
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan;
| | - Cheng-Hsin Lee
- Department of Biomedical Science and Environmental Biology, College of Life Science, Kaohsiung Medical University, Kaohsiung 80708, Taiwan; (P.-F.L.); or (C.-H.L.)
| | - Huei-Cin Sie
- Department of Pathology and Laboratory Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan;
| | - Huei-Han Liou
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan or (H.-H.L.); (L.-P.G.)
| | - Jiin-Tsuey Cheng
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (J.-T.C.); (C.-L.C.)
| | - Luo-Ping Ger
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan or (H.-H.L.); (L.-P.G.)
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; (J.-T.C.); (C.-L.C.)
| | - Chien-Chou Chen
- Family Medicine Division, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 81342, Taiwan
- Correspondence: (C.-C.C.); or (C.-F.C.); Tel.: +886-07-581-7121 (C.-C.C.); +886-07-346-8080 (C.-F.C.)
| | - Chun-Feng Chen
- Department of Stomatology, Kaohsiung Veterans General Hospital, Kaohsiung 81362, Taiwan
- Department of Dental Technology, Shu-Zen Junior College of Medicine and Management, Kaohsiung 82144, Taiwan
- School of Dentistry, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
- Correspondence: (C.-C.C.); or (C.-F.C.); Tel.: +886-07-581-7121 (C.-C.C.); +886-07-346-8080 (C.-F.C.)
| |
Collapse
|
45
|
Hapidah H, Djabir YY, Prihantono P. Increased aldehyde dehydrogenase 1 (ALDH1) levels are associated with chemo-responsiveness in breast cancer patients treated with taxane-adriamycin-cyclophosphamide regimen. Breast Dis 2021; 40:S33-S37. [PMID: 34057116 DOI: 10.3233/bd-219005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Increased plasma aldehyde dehydrogenase 1 (ALDH1) levels have been proposed to predict cancer chemoresistance. However, studies have reported inconsistent results, depending on the type of cancer cells used. OBJECTIVE This study aimed to investigate the correlation between plasma levels of ALDH1 and chemotherapy responses to the taxane-adriamycin-cyclophosphamide (TAC) regimen in breast cancer patients. METHODS Thirty breast cancer patients who underwent chemotherapy using the TAC regimen were included in this study. Blood sampling was performed before chemotherapy was initiated and after the first and third cycles of chemotherapy administration. After 3 cycles of chemotherapy, patients were categorized as non-responsive if the tumor size was reduced <30%, if the tumor size remained the same or increased, or if any new tumors were discovered. Patients were defined as responsive after 3 cycles of chemotherapy if the tumor mass disappeared, if the tumor size was reduced by at least 30% of the initial size and if no new tumors were found. RESULTS Among the 30 patients, only five were responsive to the TAC regimen. The clinical response to TAC was not correlated with the patient's age, cancer grading, or tumor stage. A change in the ALDH1 levels was observed after the third cycle of TAC administration, with significantly higher ALDH1 levels observed in responsive compared with non-responsive patients (p < 0.05). CONCLUSION The results of this study may indicate a role for ALDH1 in chemoresponsiveness, rather than chemoresistance, for the TAC regimen in breast cancer patients. Further research remains necessary to confirm this result.
Collapse
Affiliation(s)
- Hapidah Hapidah
- Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Yulia Yusrini Djabir
- Laboratory of Clinical Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Prihantono Prihantono
- Department of Surgery, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
46
|
Liskova A, Samec M, Koklesova L, Brockmueller A, Zhai K, Abdellatif B, Siddiqui M, Biringer K, Kudela E, Pec M, Gadanec LK, Šudomová M, Hassan STS, Zulli A, Shakibaei M, Giordano FA, Büsselberg D, Golubnitschaja O, Kubatka P. Flavonoids as an effective sensitizer for anti-cancer therapy: insights into multi-faceted mechanisms and applicability towards individualized patient profiles. EPMA J 2021; 12:155-176. [PMID: 34025826 PMCID: PMC8126506 DOI: 10.1007/s13167-021-00242-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 02/07/2023]
Abstract
Cost-efficacy of currently applied treatments is an issue in overall cancer management challenging healthcare and causing tremendous economic burden to societies around the world. Consequently, complex treatment models presenting concepts of predictive diagnostics followed by targeted prevention and treatments tailored to the personal patient profiles earn global appreciation as benefiting the patient, healthcare economy, and the society at large. In this context, application of flavonoids as a spectrum of compounds and their nano-technologically created derivatives is extensively under consideration, due to their multi-faceted anti-cancer effects applicable to the overall cost-effective cancer management, primary, secondary, and even tertiary prevention. This article analyzes most recently updated data focused on the potent capacity of flavonoids to promote anti-cancer therapeutic effects and interprets all the collected research achievements in the frame-work of predictive, preventive, and personalized (3P) medicine. Main pillars considered are: - Predictable anti-neoplastic, immune-modulating, drug-sensitizing effects; - Targeted molecular pathways to improve therapeutic outcomes by increasing sensitivity of cancer cells and reversing their resistance towards currently applied therapeutic modalities.
Collapse
Affiliation(s)
- Alena Liskova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Marek Samec
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Lenka Koklesova
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Aranka Brockmueller
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany
| | - Kevin Zhai
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, 24144 Doha, Qatar
| | - Basma Abdellatif
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, 24144 Doha, Qatar
| | - Manaal Siddiqui
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, 24144 Doha, Qatar
| | - Kamil Biringer
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Erik Kudela
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Martin Pec
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Laura Kate Gadanec
- Institute for Health and Sport, Victoria University, Melbourne, 3030 Australia
| | - Miroslava Šudomová
- Museum of Literature in Moravia, Klášter 1, 66461 Rajhrad, Czech Republic
| | - Sherif T. S. Hassan
- Department of Applied Ecology, Faculty of Environmental Sciences, Czech University of Life Sciences Prague, Kamýcká 129, 16500 Prague, Czech Republic
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne, 3030 Australia
| | - Mehdi Shakibaei
- Musculoskeletal Research Group and Tumor Biology, Chair of Vegetative Anatomy, Institute of Anatomy, Faculty of Medicine, Ludwig-Maximilian-University Munich, 80336 Munich, Germany
| | - Frank A. Giordano
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Bonn, Germany
| | - Dietrich Büsselberg
- Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, 24144 Doha, Qatar
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| |
Collapse
|
47
|
Proteomics of resistance to Notch1 inhibition in acute lymphoblastic leukemia reveals targetable kinase signatures. Nat Commun 2021; 12:2507. [PMID: 33947863 PMCID: PMC8097059 DOI: 10.1038/s41467-021-22787-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 03/29/2021] [Indexed: 01/01/2023] Open
Abstract
Notch1 is a crucial oncogenic driver in T-cell acute lymphoblastic leukemia (T-ALL), making it an attractive therapeutic target. However, the success of targeted therapy using γ-secretase inhibitors (GSIs), small molecules blocking Notch cleavage and subsequent activation, has been limited due to development of resistance, thus restricting its clinical efficacy. Here, we systematically compare GSI resistant and sensitive cell states by quantitative mass spectrometry-based phosphoproteomics, using complementary models of resistance, including T-ALL patient-derived xenografts (PDX) models. Our datasets reveal common mechanisms of GSI resistance, including a distinct kinase signature that involves protein kinase C delta. We demonstrate that the PKC inhibitor sotrastaurin enhances the anti-leukemic activity of GSI in PDX models and completely abrogates the development of acquired GSI resistance in vitro. Overall, we highlight the potential of proteomics to dissect alterations in cellular signaling and identify druggable pathways in cancer. NOTCH1 is a driver of T-cell acute lymphoblastic leukemia that can be inhibited by γ-secretase inhibitors (GSIs), but their clinical efficacy is limited. Here, the authors compare the phosphoproteomes of GSI resistant and sensitive models, and identify potential kinase targets to overcome GSI resistance.
Collapse
|
48
|
Garibay-Cerdenares OL, Sánchez-Meza LV, Encarnación-Guevara S, Hernández-Ortíz M, Martínez-Batallar G, Torres-Rojas FI, Mendoza-Catalán MÁ, Moral-Hernández OD, Leyva-Vázquez MA, Illades-Aguiar B. Effect of HPV 16 E6 Oncoprotein Variants on the Alterations of the Proteome of C33A Cells. Cancer Genomics Proteomics 2021; 18:273-283. [PMID: 33893080 PMCID: PMC8126335 DOI: 10.21873/cgp.20258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/19/2021] [Accepted: 02/23/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND/AIM The E6 genotypic variants of HPV 16 identified in lesions of women with cervical cancer (CC) in Southern of Mexico include the E-G350, AAa, AAc, E-C188/G350, and E-A176/G350, transcriptomic analysis cells transfected with those variants showed to induce differential expression of the host genes involved in the development of CC, the aim of this work was to understand how the over-expression of the E6 oncoprotein and its variants can induce molecular mechanisms that lead to more aggressive HPV 16 phenotypes in cervical cancer and which proteins could be associated with the process. MATERIALS AND METHODS Total extracts from C33A, C33A mock, C33A AAa, C33A E-C188/G350, C33A E-A176/G350, and C33A E-prototype cells were analyzed using 2D electrophoresis, PDQuest software and mass spectrometry, validation of results was performed through qPCR. RESULTS Statistically significant differential expression of 122 spots was detected, 12 of the identified proteins were associated with metabolism and metabolic programming. Out of these CCT8, ENO and ALDH1A were further validated. CONCLUSION CCT8 and ALDH1A were found to be over-expressed in C33A AAa and C33A E-A176/G350, compared to the E prototype. Both proteins could be associated with a most aggressive phenotype due to their relationship with metabolism, protein folding and stemness, mechanisms associated to E6 that could be useful in the design of new therapies.
Collapse
Affiliation(s)
- Olga Lilia Garibay-Cerdenares
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, México
- CONACyT- Universidad Autónoma de Guerrero, Chilpancingo, México
| | - Luz Victoria Sánchez-Meza
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, México
| | | | | | | | - Francisco Israel Torres-Rojas
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, México
| | - Miguel Ángel Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, México
| | - Oscar Del Moral-Hernández
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, México
| | - Marco Antonio Leyva-Vázquez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, México
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, México;
| |
Collapse
|
49
|
Cermeño EA, O'Melia MJ, Han WM, Veith A, Barber G, Huang EH, Thomas SN, García AJ. Hydrodynamic shear-based purification of cancer cells with enhanced tumorigenic potential. Integr Biol (Camb) 2021; 12:1-11. [PMID: 31965190 DOI: 10.1093/intbio/zyz038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 11/01/2019] [Accepted: 11/05/2019] [Indexed: 11/13/2022]
Abstract
Tumor-initiating cells (TICs), a subpopulation of cancerous cells with high tumorigenic potential and stem-cell-like properties, drive tumor progression and are resistant to conventional therapies. Identification and isolation of TICs are limited by their low frequency and lack of robust markers. Here, we characterize the heterogeneous adhesive properties of a panel of human and murine cancer cells and demonstrate differences in adhesion strength among cells, which exhibit TIC properties and those that do not. These differences in adhesion strength were exploited to rapidly (~10 min) and efficiently isolate cancerous cells with increased tumorigenic potential in a label-free manner by use of a microfluidic technology. Isolated murine and human cancer cells gave rise to larger tumors with increased growth rate and higher frequency in both immunocompetent and immunocompromised mice, respectively. This rapid and label-free TIC isolation technology has the potential to be a valuable tool for facilitating research into TIC biology and the development of more efficient diagnostics and cancer therapies.
Collapse
Affiliation(s)
- Efraín A Cermeño
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Meghan J O'Melia
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA.,Coulter Department of Biomedical Engineering, Georgia Tech/Emory, Atlanta, GA, USA
| | - Woojin M Han
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Austin Veith
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Graham Barber
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Emina H Huang
- Lerner Research Institute, Department of Cancer Biology, Department of Colorectal Surgery, Cleveland Clinic, Cleveland, OH, USA
| | - Susan N Thomas
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.,Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| |
Collapse
|
50
|
Wojtowicz K, Sterzyńska K, Świerczewska M, Nowicki M, Zabel M, Januchowski R. Piperine Targets Different Drug Resistance Mechanisms in Human Ovarian Cancer Cell Lines Leading to Increased Sensitivity to Cytotoxic Drugs. Int J Mol Sci 2021; 22:ijms22084243. [PMID: 33921897 PMCID: PMC8073496 DOI: 10.3390/ijms22084243] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 01/20/2023] Open
Abstract
Our goal was to examine the anticancer effects of piperine against the resistant human ovarian cancer cells and to explore the molecular mechanisms responsible for its anticancer effects. Our study used drug-sensitive ovarian cancer cell line W1 and its sublines resistant to paclitaxel (PAC) and topotecan (TOP). We analyzed the cytotoxic effect of piperine and cytostatic drugs using an MTT assay. The impact of piperine on protein expression was determined by immunofluorescence and Western blot. We also examined its effect on cell proliferation and migration. We noticed a different level of piperine resistance between cell lines. Piperine increases the cytotoxic effect of PAC and TOP in drug-resistant cells. We observed an increase in PTPRK expression correlated with decreased pTYR level after piperine treatment and downregulation of P-gp and BCRP expression. We also noted a decrease in COL3A1 and TGFBI expression in investigated cell lines and increased COL3A1 expression in media from W1PR2 cells. The expression of Ki67 protein and cell proliferation rate decreased after piperine treatment. Piperine markedly inhibited W1TR cell migration. Piperine can be considered a potential anticancer agent that can increase chemotherapy effectiveness in cancer patients.
Collapse
Affiliation(s)
- Karolina Wojtowicz
- Department of Histology and Embryology, Poznań University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland; (K.S.); (M.Ś.); (M.N.)
- Correspondence: (K.W.); (R.J.)
| | - Karolina Sterzyńska
- Department of Histology and Embryology, Poznań University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland; (K.S.); (M.Ś.); (M.N.)
| | - Monika Świerczewska
- Department of Histology and Embryology, Poznań University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland; (K.S.); (M.Ś.); (M.N.)
| | - Michał Nowicki
- Department of Histology and Embryology, Poznań University of Medical Sciences, Święcickiego 6 St., 61-781 Poznań, Poland; (K.S.); (M.Ś.); (M.N.)
| | - Maciej Zabel
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, Zyty 28 St., 65-046 Zielona Gora, Poland;
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, T. Chałubińskiego 6a St., 50-368 Wroclaw, Poland
| | - Radosław Januchowski
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, Zyty 28 St., 65-046 Zielona Gora, Poland;
- Correspondence: (K.W.); (R.J.)
| |
Collapse
|