1
|
Muñoz-Osses M, Navarrete E, Morales P, Quiroz J, Silva M, Torres-González S, Vásquez-Martínez Y, Godoy F, Mascayano C. Substituted aryl piperazine ligands as new dual 5-hLOX/COX-2 inhibitors. Synthesis, biological and computational studies. Bioorg Chem 2025; 159:108398. [PMID: 40174530 DOI: 10.1016/j.bioorg.2025.108398] [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: 01/13/2025] [Revised: 03/10/2025] [Accepted: 03/18/2025] [Indexed: 04/04/2025]
Abstract
Two series of cyano (1a-l) and amino (2a-l) aryl piperazines were synthesized and evaluated for their inhibitory activity against 5-lipoxygenase (5-hLOX) and cyclooxygenase-2 (COX-2). The newly designed derivatives feature diphenyl methyl (a-d), phenyl (e-h), or methoxyphenyl (i-l) groups, respectively, and demonstrated significant inhibition of 5-hLOX. Noteworthy were compounds 1b, 1 g, 1 k, 2f, and 2 g, exhibiting IC50 values ranging from 2.2 to 3.3 μM. The most potent inhibitors (1b, 1 g, 1 k, 2c, and 2f) were characterized by a competitive inhibition mechanism, with Ki values ranging between 1.77 μM and 9.50 μM. Additionally, compounds 2a, 2b, 2 g, and 2 h displayed promising dual inhibition of 5-hLOX and COX-2, with IC50 values below 15 μM. Cytotoxicity assessments against HEK293 cells revealed that the cyano derivatives (1a-l) were non-cytotoxic (CC50 > 200 μM), whereas the amino derivatives (2a-l) exhibited moderate cytotoxicity (CC50 < 50 μM). Notably, the most active derivatives against both targets were non-cytotoxic at their respective inhibitory concentrations. Computational studies, including docking and molecular dynamics simulations, indicated that compound 1 g demonstrated greater stability within the catalytic site of 5-hLOX compared to compound 2f, correlating with the higher affinity observed in kinetic assays. Furthermore, quantitative structure-activity relationship (QSAR) analyses revealed strong correlations between theoretical and experimental IC50 values (97 % for 1a-l and 93 % for 2a-l). These findings, combined with absorption, distribution, metabolism, and excretion (ADME) predictions, suggest that these derivatives are promising candidates as dual inhibitors of 5-hLOX and COX-2.
Collapse
Affiliation(s)
| | | | - Pilar Morales
- Departamento Ciencias del Ambiente, Universidad de Santiago de Chile, Chile
| | - Javiera Quiroz
- Departamento Ciencias del Ambiente, Universidad de Santiago de Chile, Chile
| | - Maite Silva
- Departamento Química de los Materiales, Universidad de Santiago de Chile, Chile
| | | | - Yesseny Vásquez-Martínez
- Programa Centro de Investigaciones Biomédicas y Aplicadas (CIBAP), Escuela de Medicina, Facultad de Ciencias Médicas, Universidad de Santiago de Chile, Chile
| | - Fernando Godoy
- Departamento Química de los Materiales, Universidad de Santiago de Chile, Chile
| | - Carolina Mascayano
- Departamento Ciencias del Ambiente, Universidad de Santiago de Chile, Chile
| |
Collapse
|
2
|
Iqbal A, Abbas W, Ejaz S, Riaz N, Ashok AK, Hayat MM, Ashraf M. Multimodal evaluation of lipoxygenase-targeting NSAIDs using integrated in vitro, SAR, in silico, cytotoxicity towards MCF-7 cell line, DNA docking and MD simulation approaches. Int J Biol Macromol 2025; 314:143665. [PMID: 40316117 DOI: 10.1016/j.ijbiomac.2025.143665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 04/12/2025] [Accepted: 04/28/2025] [Indexed: 05/04/2025]
Abstract
Lipoxygenase (LOX) and cyclooxygenase (COX) pathways generate biologically active mediators implicated in inflammatory disorders and several classes of cancer. Non-steroidal anti-inflammatory drugs (NSAIDs) inhibit the COX pathway by inhibiting the COX-1 and COX-2 enzymes. We reported earlier that several NSAIDs, including naproxen, aspirin and acetaminophen, inhibited lipoxygenase (LOX) enzyme at sub-micromolar concentrations. In continuation, the present work demonstrates the anti-LOX activity of nine more NSAIDs supported by in vitro, in silico, MD simulation and breast cancer cell line studies. All tested drugs displayed potent to excellent inhibitory profiles with IC50 values <24.93 ± 0.64 μM. Aceclofenac (IC50 0.85 ± 0.06 μM) was the most active drug, followed by indomethacin (IC50 1.13 ± 0.07 μM), meloxicam (IC50 1.94 ± 0.07 μM) and ketorolac (IC50 9.26 ± 0.82 μM). Celecoxib (IC50 15.81 ± 0.71 μM), lornoxicam (IC50 16.54 ± 0.28 μM) and nimesulide (IC50 19.87 ± 0.85 μM) showed excellent inhibitory profiles. Flurbiprofen (IC50 21.73 ± 0.93 μM) and etoricoxib (IC50 24.93 ± 0.64 μM) moderately inhibited the target enzyme. SAR studies revealed that active molecules decorated with the carboxylate group afforded strong binding interactions as observed by in vitro assays and structural features. Other drugs, including enol derivatives and celecoxib, also showcased enhanced binding interactions. However, the cytotoxic effects of NSAIDs against the MCF-7 breast cancer cell line did not disclose significant anticancer activity. Molecular docking studies against human 5-LOX offered the best binding affinities for aceclofenac (-13.54 kcal/mol), accompanied by conventional hydrogen bonding and hydrophobic interactions as supported by the in vitro results. Docking studies with DNA dodecamer established minor groove binding with their possible role in DNA replication and gene expression. Density functional theory (DFT) and ESP studies, MD simulations and MMPBSA free energy calculations further reiterated the stability of ligand-receptor complexes. Overall, these findings highlight the potential of targeted NSAIDs as dual COX/LOX inhibitors with broader therapeutic relevance in inflammatory disorders.
Collapse
Affiliation(s)
- Ambar Iqbal
- Institute of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan; Department of Biochemistry, Institute of Biochemistry, Biotechnology & Bioinformatics (IBBB), B.J. Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan.
| | - Wasim Abbas
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering College, Pakistan Institute of Engineering and Applied Sciences (NIBGE-C, PIEAS), Faisalabad, Punjab, Pakistan
| | - Samina Ejaz
- Department of Biochemistry, Institute of Biochemistry, Biotechnology & Bioinformatics (IBBB), B.J. Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Naheed Riaz
- Institute of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan.
| | - Avinash Karkada Ashok
- Department of Biotechnology, Siddaganga Institute of Technology, Tumakuru, Karnataka 572103, India
| | | | - Muhammad Ashraf
- Institute of Chemistry, Baghdad-ul-Jadeed Campus, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan.
| |
Collapse
|
3
|
Mandle HB, Jenab M, Gunter MJ, Tjønneland A, Olsen A, Dahm CC, Zhang J, Sugier PE, Rothwell J, Severi G, Kaaks R, Katzke VA, Schulze MB, Masala G, Sieri S, Panico S, Sacerdote C, Bonet C, Sánchez MJ, Amiano P, Huerta JM, Guevara M, Palmqvist R, Löwenmark T, Perez-Cornago A, Weiderpass E, Heath AK, Cross AJ, Vineis P, Hughes DJ, Fedirko V. Inflammation and gut barrier function-related genes and colorectal cancer risk in western European populations. Mutagenesis 2025; 40:48-60. [PMID: 38441165 PMCID: PMC11911009 DOI: 10.1093/mutage/geae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 04/04/2024] [Indexed: 03/21/2024] Open
Abstract
Gut barrier dysfunction and related inflammation are known to be associated with the development and progression of colorectal cancer (CRC). We investigated associations of 292 single-nucleotide polymorphisms (SNPs) from 27 genes related to endotoxins/lipopolysaccharide (LPS) sensing and tolerance, mucin synthesis, inflammation, and Crohn's disease with colon and rectal cancer risks. Incident CRC cases (N = 1374; colon = 871, rectum = 503) were matched 1:1 to controls nested within the European Prospective Investigation into Cancer and Nutrition cohort. Previously measured serum concentrations of gut barrier function and inflammation biomarkers (flagellin/LPS-specific immunoglobulins and C-reactive protein [CRP]) were available for a sub-set of participants (Ncases = 1001; Ncontrols = 667). Forty-two unique SNPs from 19 different genes were associated with serum biomarkers at Punadjusted ≤ 0.05 among controls. Among SNPs associated with a gut permeability score, 24 SNPs were in genes related to LPS sensing and mucin synthesis. Nine out of 12 SNPs associated with CRP were in genes related to inflammation or Crohn's disease. TLR4 was associated with colon cancer at the SNP level (nine SNPs, all Punadjusted ≤ 0.04) and at the gene level (Punadjusted ≤ 0.01). TLR4 rs10759934 was associated with rectal cancer but not colon cancer. Similarly, IL10 was associated with rectal cancer risk at an SNP and gene level (both Punadjusted ≤ 0.01), but not colon cancer. Genes and SNPs were selected a priori; therefore, we present unadjusted P-values. However, no association was statistically significant after multiple testing correction. This large and comprehensive study has identified gut barrier function and inflammation-related genes possibly contributing to CRC risk in European populations and is consistent with potential etiological links between host genetic background, gut barrier permeability, microbial endotoxemia, and CRC development.
Collapse
Affiliation(s)
- Hannah B Mandle
- Department of Epidemiology, Emory Rollins School of Public Health, Atlanta, GA 30322, USA
| | - Mazda Jenab
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
| | - Marc J Gunter
- Nutrition and Metabolism Branch, International Agency for Research on Cancer (IARC-WHO), 69372 Lyon, France
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, SW7 2AZ, UK
| | - Anne Tjønneland
- Diet, Cancer and Health, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark
- Department of Public Health, University of Copenhagen, 1353 Copenhagen K, Denmark
| | - Anja Olsen
- Department of Public Health, University of Copenhagen, 1353 Copenhagen K, Denmark
- Department of Public Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Christina C Dahm
- Department of Public Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Jie Zhang
- Department of Public Health, Aarhus University, DK-8000 Aarhus C, Denmark
| | - Pierre-Emmanuel Sugier
- Université Paris-Saclay, UVSQ, Inserm ‘Exposome and Heredity’ team, CESP U1018, 94807 Villejuif Cedex, France
- Laboratoire de Mathématiques et de leurs Applications de Pau E2S UPPA, CNRS, 64013 Pau Cedex, France
| | - Joseph Rothwell
- Université Paris-Saclay, UVSQ, Inserm ‘Exposome and Heredity’ team, CESP U1018, 94807 Villejuif Cedex, France
| | - Gianluca Severi
- Université Paris-Saclay, UVSQ, Inserm ‘Exposome and Heredity’ team, CESP U1018, 94807 Villejuif Cedex, France
| | - Rudolf Kaaks
- Division of Cancer Epidemiology, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Verena A Katzke
- Division of Cancer Epidemiology, German Cancer Research Center, DKFZ, 69120 Heidelberg, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition, Potsdam-Rehbruecke, 14469 Nuthetal, Germany
- Institute of Nutritional Science, University of Potsdam, 14469 Nuthetal, Germany
| | - Giovanna Masala
- Institute for Cancer Research, Prevention and Clinical Network (ISPRO), 50139 Florence, Italy
| | - Sabina Sieri
- Epidemiology and Prevention Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, 20133 Milan, Italy
| | - Salvatore Panico
- Dipartimento Di Medicina Clinica E Chirurgia, Federico II University, 80131 Naples, Italy
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, AOU Città della Salute e della Scienza University Hospital, 10126 Turin, Italy
| | - Catalina Bonet
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program, Catalan Institute of Oncology (ICO), L’Hospitalet de Llobregat, 0890x Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), L’Hospitalet de Llobregat, 0890x Barcelona, Spain
| | - Maria-Jose Sánchez
- Escuela Andaluza de Salud Pública (EASP), 18011 Granada, Spain
- Instituto de Investigación Biosanitaria ibs.18011 Granada, Spain
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Department of Preventive Medicine and Public Health, University of Granada, 18071 Granada, Spain
| | - Pilar Amiano
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Ministry of Health of the Basque Government, Sub Directorate for Public Health and Addictions of Gipuzkoa, BioGipuzkoa Health Research Institute, Epidemiology of Chronic and Communicable Diseases Group, 20014 Donostia – San Sebastian, Spain
| | - José María Huerta
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Department of Epidemiology, Murcia Regional Health Council-IMIB, 30120, El Palmar, Murcia, Spain
| | - Marcela Guevara
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Instituto de Salud Pública y Laboral de Navarra, 31003 Pamplona, Navarra, Spain
- Navarra Institute for Health Research (IdiSNA), 31008 Pamplona, Navarra, Spain
| | - Richard Palmqvist
- Department of Medical Biosciences, Umea University, 901 87 Umeå, Sweden
| | - Thyra Löwenmark
- Department of Medical Biosciences, Umea University, 901 87 Umeå, Sweden
| | - Aurora Perez-Cornago
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford OX3 7LF, UK
| | - Elisabete Weiderpass
- Office of the Director, International Agency for Research on Cancer, 69366 Lyon Cedex 07, France
| | - Alicia K Heath
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, SW7 2AZ, UK
| | - Amanda J Cross
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, SW7 2AZ, UK
| | - Paolo Vineis
- MRC Centre for Environment and Health, School of public Health, Imperial College London, London W2 1PG, UK
- Italian Institute for Genomic Medicine (IIGM), 10060 Candiolo TO,Italy
| | - David J Hughes
- Cancer Biology and Therapeutics Group, School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Veronika Fedirko
- Department of Epidemiology, Emory Rollins School of Public Health, Atlanta, GA 30322, USA
- Department of Epidemiology, University of Texas M. D. Anderson Cancer Center, 77030 Houston, TX, USA
| |
Collapse
|
4
|
Lu W, Aihaiti A, Abudukeranmu P, Liu Y, Gao H. Arachidonic acid metabolism as a novel pathogenic factor in gastrointestinal cancers. Mol Cell Biochem 2025; 480:1225-1239. [PMID: 38963615 DOI: 10.1007/s11010-024-05057-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Gastrointestinal (GI) cancers are a major global health burden, representing 20% of all cancer diagnoses and 22.5% of global cancer-related deaths. Their aggressive nature and resistance to treatment pose a significant challenge, with late-stage survival rates below 15% at five years. Therefore, there is an urgent need to delve deeper into the mechanisms of gastrointestinal cancer progression and optimize treatment strategies. Increasing evidence highlights the active involvement of abnormal arachidonic acid (AA) metabolism in various cancers. AA is a fatty acid mainly metabolized into diverse bioactive compounds by three enzymes: cyclooxygenase, lipoxygenase, and cytochrome P450 enzymes. Abnormal AA metabolism and altered levels of its metabolites may play a pivotal role in the development of GI cancers. However, the underlying mechanisms remain unclear. This review highlights a unique perspective by focusing on the abnormal metabolism of AA and its involvement in GI cancers. We summarize the latest advancements in understanding AA metabolism in GI cancers, outlining changes in AA levels and their potential role in liver, colorectal, pancreatic, esophageal, gastric, and gallbladder cancers. Moreover, we also explore the potential of targeting abnormal AA metabolism for future therapies, considering the current need to explore AA metabolism in GI cancers and outlining promising avenues for further research. Ultimately, such investigations aim to improve treatment options for patients with GI cancers and pave the way for better cancer management in this area.
Collapse
Affiliation(s)
- Weiqin Lu
- General Surgery, Cancer Center, Department of Vascular Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | | | | | - Yajun Liu
- Aksu First People's Hospital, Xinjiang, China
| | - Huihui Gao
- Cancer Center, Department of Hospital Infection Management and Preventive Medicine, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Ricci MF, Lourenço EMG, Pereira RDD, Araújo RRS, Oliveira FBR, Barbosa da Silva E, de Oliveira GS, Teixeira MM, Rocha NDN, Chambergo FS, Roman-Campos D, Cruz JS, Ferreira RS, Machado FS. Zileuton, a 5-Lypoxigenase Inhibitor, is Antiparasitic and Prevents Inflammation in the Chronic Stage of Heart Chagas Disease. ACS Infect Dis 2024; 10:4258-4270. [PMID: 39609255 DOI: 10.1021/acsinfecdis.4c00623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Chronic Chagas cardiomyopathy is associated with an unbalanced immune response and impaired heart function, and available drugs do not prevent its development. Zileuton (Zi), a 5-lypoxigenase inhibitor, affects inflammatory/pro-resolution mediators. Herein, Zi treatment in the early phase of infection reduced parasitemia associated mainly with the direct effect of Zi on the parasite, and the enzyme epoxide hydrolase was the potential molecular target behind the trypanocidal effect. In the intermediate acute phase of infection, Zi reduced the number of innate and adaptive inflammatory cells, increased the level of SOCS2 expression in the heart associated with lower inflammation, and improved cardiac function. Zi treatment initiated in the chronic stage increased the level of SOCS2 expression in the heart, reduced inflammation, and improved cardiac function. Our data suggest that Zi protects against Trypanosoma cruzi infection by acting directly on the parasite and reducing heart damage and is a promising option for the treatment of Chagas disease.
Collapse
Affiliation(s)
- Mayra Fernanda Ricci
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Estela Mariana Guimarães Lourenço
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Rafaela das Dores Pereira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Ronan Ricardo Sabino Araújo
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Fernando Bento Rodrigues Oliveira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Elany Barbosa da Silva
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Gabriel Stephani de Oliveira
- Department of Microbiology, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo 05508-000, Brazil
| | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
- Program in Health Sciences: Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Minas Gerais, Brazil
| | - Nazareth de Novaes Rocha
- Department of Physiology and Pharmacology, Biomedical Institute, Universidade Federal Fluminense, Niterói 24020-141, Rio de Janeiro, Brazil
| | | | - Danilo Roman-Campos
- Department of Biophysics, Universidade Federal de São Paulo, São Paulo 04023-062, Brazil
| | - Jader Santos Cruz
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Rafaela Salgado Ferreira
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Fabiana Simão Machado
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
- Program in Health Sciences: Infectious Diseases and Tropical Medicine/Interdisciplinary Laboratory of Medical Investigation, Faculty of Medicine, Universidade Federal de Minas Gerais, Belo Horizonte 30130-100, Minas Gerais, Brazil
| |
Collapse
|
6
|
Bošković J, Dobričić V, Savić J, Rupar J, Aleksić M, Marković B, Čudina O. In Vitro Evaluation of Pharmacokinetic Properties of Selected Dual COX-2 and 5-LOX Inhibitors. Pharmaceuticals (Basel) 2024; 17:1329. [PMID: 39458971 PMCID: PMC11510591 DOI: 10.3390/ph17101329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/28/2024] Open
Abstract
Evaluation of pharmacokinetic properties is a significant step at the early stages of drug development. In this study, an in vitro evaluation of the pharmacokinetic properties of five newly synthesized compounds was performed. These compounds belong to N-hydroxyurea and hydroxamic acid derivatives and analogs of NSAIDs indomethacin, flurbiprofen, diclofenac, ibuprofen, and naproxen (compounds 1, 2, 3, 11, and 12, respectively) with dual COX-2 and 5-LOX inhibitory activity. Two in vitro methods (biopartitioning micellar chromatography (BMC) and PAMPA) were used to evaluate passive gastrointestinal absorption, while high-performance affinity chromatography (HPAC) and differential pulse voltammetry (DPV) were used to evaluate binding to human serum albumin (HSA). The introduction of N-hydroxyurea and hydroxamic acid groups into the structure of NSAIDs decreases both expected passive gastrointestinal absorption (BMC k values were from 3.02 to 9.50, while for NSAIDs were from 5.29 to 13.36; PAMPA -logPe values were between 3.81 and 4.76, while for NSAIDs were ≤3.46) and HSA binding (HPAC logk values were from 2.03 to 9.54, while for NSAIDs were ≥11.03; DPV peak potential shifts were between 7 and 34, while for NSAIDs were ≥54). Structural modifications of all tested compounds that increase lipophilicity could be considered to enhance their passive gastrointestinal absorption. Considering lower expected HSA binding and higher lipophilicity of tested compounds compared to corresponding NSAIDs, it can be expected that the volume of distribution of compounds 1, 2, 3, 11, and 12 will be higher. Reduced HSA binding may also decrease interactions with other drugs in comparison to corresponding NSAIDs. All tested compounds showed significant microsomal instability (25.07-58.44% decrease in concentration) in comparison to indomethacin (14.47%) and diclofenac (20.99%).
Collapse
Affiliation(s)
- Jelena Bošković
- Department of Pharmaceutical Chemistry, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia (B.M.)
| | - Vladimir Dobričić
- Department of Pharmaceutical Chemistry, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia (B.M.)
| | - Jelena Savić
- Department of Pharmaceutical Chemistry, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia (B.M.)
| | - Jelena Rupar
- Department of Physical Chemistry and Instrumental Methods, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Mara Aleksić
- Department of Physical Chemistry and Instrumental Methods, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Bojan Marković
- Department of Pharmaceutical Chemistry, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia (B.M.)
| | - Olivera Čudina
- Department of Pharmaceutical Chemistry, University of Belgrade–Faculty of Pharmacy, Vojvode Stepe 450, 11000 Belgrade, Serbia (B.M.)
| |
Collapse
|
7
|
Amoah AS, Pestov NB, Korneenko TV, Prokhorenko IA, Kurakin GF, Barlev NA. Lipoxygenases at the Intersection of Infection and Carcinogenesis. Int J Mol Sci 2024; 25:3961. [PMID: 38612771 PMCID: PMC11011848 DOI: 10.3390/ijms25073961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/08/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
The persisting presence of opportunistic pathogens like Pseudomonas aeruginosa poses a significant threat to many immunocompromised cancer patients with pulmonary infections. This review highlights the complexity of interactions in the host's defensive eicosanoid signaling network and its hijacking by pathogenic bacteria to their own advantage. Human lipoxygenases (ALOXs) and their mouse counterparts are integral elements of the innate immune system, mostly operating in the pro-inflammatory mode. Taking into account the indispensable role of inflammation in carcinogenesis, lipoxygenases have counteracting roles in this process. In addition to describing the structure-function of lipoxygenases in this review, we discuss their roles in such critical processes as cancer cell signaling, metastases, death of cancer and immune cells through ferroptosis, as well as the roles of ALOXs in carcinogenesis promoted by pathogenic infections. Finally, we discuss perspectives of novel oncotherapeutic approaches to harness lipoxygenase signaling in tumors.
Collapse
Affiliation(s)
- Abdul-Saleem Amoah
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Laboratory of Molecular Oncology, Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny 141701, Russia
| | - Nikolay B. Pestov
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Moscow 108819, Russia
- Vavilov Institute of General Genetics, Moscow 119991, Russia
| | - Tatyana V. Korneenko
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
| | - Igor A. Prokhorenko
- Group of Cross-Linking Enzymes, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow 117997, Russia; (T.V.K.); (I.A.P.)
| | - Georgy F. Kurakin
- Department of Biochemistry, Pirogov Russian National Research Medical University, Moscow 117513, Russia;
| | - Nickolai A. Barlev
- Institute of Biomedical Chemistry, Moscow 119121, Russia; (A.-S.A.); (N.A.B.)
- Laboratory of Tick-Borne Encephalitis and Other Viral Encephalitides, Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products, Moscow 108819, Russia
| |
Collapse
|
8
|
Meng YW, Liu JY. Pathological and pharmacological functions of the metabolites of polyunsaturated fatty acids mediated by cyclooxygenases, lipoxygenases, and cytochrome P450s in cancers. Pharmacol Ther 2024; 256:108612. [PMID: 38369063 DOI: 10.1016/j.pharmthera.2024.108612] [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: 10/30/2023] [Revised: 01/19/2024] [Accepted: 02/05/2024] [Indexed: 02/20/2024]
Abstract
Oxylipins have garnered increasing attention because they were consistently shown to play pathological and/or pharmacological roles in the development of multiple cancers. Oxylipins are the metabolites of polyunsaturated fatty acids via both enzymatic and nonenzymatic pathways. The enzymes mediating the metabolism of PUFAs include but not limited to lipoxygenases (LOXs), cyclooxygenases (COXs), and cytochrome P450s (CYPs) pathways, as well as the down-stream enzymes. Here, we systematically summarized the pleiotropic effects of oxylipins in different cancers through pathological and pharmacological aspects, with specific reference to the enzyme-mediated oxylipins. We discussed the specific roles of oxylipins on cancer onset, growth, invasion, and metastasis, as well as the expression changes in the associated metabolic enzymes and the associated underlying mechanisms. In addition, we also discussed the clinical application and potential of oxylipins and related metabolic enzymes as the targets for cancer prevention and treatment. We found the specific function of most oxylipins in cancers, especially the underlying mechanisms and clinic applications, deserves and needs further investigation. We believe that research on oxylipins will provide not only more therapeutic targets for various cancers but also dietary guidance for both cancer patients and healthy humans.
Collapse
Affiliation(s)
- Yi-Wen Meng
- CNTTI of the Institute of Life Sciences & Department of Anesthesia of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing 400016, China
| | - Jun-Yan Liu
- CNTTI of the Institute of Life Sciences & Department of Anesthesia of the Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China; Basic Medicine Research and Innovation Center for Novel Target and Therapeutic Intervention, Ministry of Education, Chongqing 400016, China; College of Pharmacy, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
9
|
Chen J, Tang Y, Qin D, Yu X, Tong H, Tang C, Tang Z. ALOX5 acts as a key role in regulating the immune microenvironment in intrahepatic cholangiocarcinoma, recruiting tumor-associated macrophages through PI3K pathway. J Transl Med 2023; 21:923. [PMID: 38124204 PMCID: PMC10734103 DOI: 10.1186/s12967-023-04804-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Intrahepatic cholangiocarcinoma (ICC) is poorly treated due to the presence of an inhibitory immune microenvironment. Tumor-associated macrophages (TAM) are an important component of TME. ALOX5 is an important lipid metabolism enzyme in cancer progression, but the mechanism by which it regulates TAM to promote ICC progression is unknown. The aim of this study was to investigate the potential mechanism of TAM regulation by ALOX5 and the translational effect of targeting ALOX5. METHODS In this study, we investigated the association between the spatial localization of epithelial cells and TAMs by combining scRNA-seq analysis with multiplex immunofluorescence analysis. Through bulk sequencing analysis and spatial analysis, lipid metabolism genes closely related to TAM infiltration were screened. In vitro co-culture model was constructed to verify that ALOX5 and its downstream metabolite LTB4 promote M2 macrophage migration. Bulk sequencing after co-culture combined with single-cell analysis was performed to identify key pathways for up-regulation of M2 macrophage migration. Finally, the effect of CSF1R inhibitor (PLX3397) combined with ALOX5 inhibitor (Zileuton) in vivo was investigated by by xenograft tumor formation experiment in nude mice. RESULTS ALOX5 in ICC cells was a key lipid metabolism gene affecting the infiltration of M2 macrophages in TME. Mechanically, LTB4, a metabolite downstream of ALOX5, recruited M2 macrophages to migrate around tumor cells by binding to BLT1/BLT2 and activating the PI3K pathway, which ultimately lead to the promotion of ICC progression. Targeting CSF1R in combination with ALOX5 inhibitor effectively reduced tumor volume and M2 macrophage infiltration abundance. CONCLUSION In ICC, LTB4, a metabolite secreted by ALOX5 of epithelial cells, binded to BLT1/BLT2 on TAM surface to activate PI3K pathway and promote TAM migration, thus promoting ICC progression. Targeting CSF1R in combination with ALOX5 inhibitor for ICC is a promising combination therapy modality.
Collapse
Affiliation(s)
- Jialu Chen
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yue Tang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Delong Qin
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Xiaopeng Yu
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Huanjun Tong
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Chengwei Tang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China
| | - Zhaohui Tang
- Department of General Surgery, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China.
- Department of Blood Transfusion, Xinhua Hospital, Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, China.
- Shanghai Key Laboratory of Biliary Tract Disease Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
| |
Collapse
|
10
|
Kizir D, Karaman M, Ceylan H. Tannic acid may ameliorate doxorubicin-induced changes in oxidative stress parameters in rat spleen. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:3605-3613. [PMID: 37272930 DOI: 10.1007/s00210-023-02563-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 05/30/2023] [Indexed: 06/06/2023]
Abstract
Doxorubicin (DOX) is a potent and broad-spectrum drug widely used in the treatment of cancer. However, the toxicity and side effects of DOX on various organs limit its clinical use. Approaches using natural antioxidants with these drugs have the potential to alleviate negative side effects. The aim of this study was to investigate the potential protective effect of tannic acid, a polyphenolic compound found naturally in plants, against DOX-induced spleen toxicity. Expression levels of Alox5, Inos, IL-6, Tnf-α, Casp-3, Bax, SOD, GST, CAT and GPx genes were determined using cDNAs obtained from spleen tissues of rats treated with DOX, tannic acid and both. In addition, SOD, CAT, GPx and GST enzyme activities, and GSH and MDA levels were measured in tissues. In the spleen tissues, DOX caused a decrease in the level of GSH and an increase in the level of MDA. In addition, it was determined that DOX had a suppressive effect on CAT, GST, SOD and GPx mRNA levels and its enzyme activities, which are antioxidant system components. The mRNA expression levels of proinflammatory cytokine markers, apoptotic genes, and some factors involved in cell metabolism showed a change compared to the control after DOX application. However, as a result of tannic acid treatment with DOX, these changes approached the values of the control group. The findings showed that tannic acid had a protective effect on the changes in the oxidative stress and inflammation system in the rat spleen as a result of the application of tannic acid together with DOX.
Collapse
Affiliation(s)
- Duygu Kizir
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, 25240, Erzurum, Turkey
| | - Melike Karaman
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, 25240, Erzurum, Turkey.
| | - Hamid Ceylan
- Department of Molecular Biology and Genetics, Faculty of Science, Atatürk University, 25240, Erzurum, Turkey
| |
Collapse
|
11
|
Hu WM, Liu SQ, Zhu KF, Li W, Yang ZJ, Yang Q, Zhu ZC, Chang J. The ALOX5 inhibitor Zileuton regulates tumor-associated macrophage M2 polarization by JAK/STAT and inhibits pancreatic cancer invasion and metastasis. Int Immunopharmacol 2023; 121:110505. [PMID: 37348233 DOI: 10.1016/j.intimp.2023.110505] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/24/2023] [Accepted: 06/12/2023] [Indexed: 06/24/2023]
Abstract
5-lipoxygenase (encoded by ALOX5) plays an important role in immune regulation. Zileuton is currently the only approved ALOX5 inhibitor. However, the mechanisms of ALOX5 and Zileuton in progression of pancreatic cancer remain unclear. Therefore, we investigated the effects of Zileuton on tumor-associated macrophage M2 polarization and pancreatic cancer invasion and metastasis, both in vivo and in vitro. In bulk RNA sequencing (RNA-seq) and single-cell RNA sequencing (scRNA-seq) analyses, we found a significant association between elevated levels of ALOX5 and poor survival, adverse stages, M2 macrophage infiltration, and the activation of JAK/STAT pathways in macrophages. In clinical samples, immunofluorescence, quantitative real-time PCR and immunohistochemical results verified the high expression of ALOX5 in pancreatic cancer, primarily in macrophages. We constructed PANC-1 human pancreatic cancer cells and macrophages overexpressing ALOX5 using lentivirus. In PANC-1 pancreatic cancer cells, low-dose Zileuton inhibited PANC-1 cell invasion and migration by blocking ALOX5. In macrophages, ALOX5 induced the M2-like phenotype through the JAK/STAT pathway and promoted the chemotaxis of macrophages towards PANC-1 cells, while Zileuton can inhibit these effects. We constructed the nude mouse model of in situ transplantation tumor of pancreatic cancer. After treatment with Zileuton, the mice showed increased survival rates and reduced liver metastasis. These findings indicate that ALOX5 regulates tumor-associated macrophage M2 polarization via the JAK/STAT pathway and promotes invasion and metastasis in pancreatic cancer. Zileuton can inhibit these effects by inhibiting ALOX5. These results provide a theoretical basis for the potential use of Zileuton in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Wei-Min Hu
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Si-Qing Liu
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Kong-Fan Zhu
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Wei Li
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Zhi-Jian Yang
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Qiang Yang
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China
| | - Zhong-Chao Zhu
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China.
| | - Jian Chang
- Department of Pancreatic Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, PR China.
| |
Collapse
|
12
|
Ortiz-Placín C, Castillejo-Rufo A, Estarás M, González A. Membrane Lipid Derivatives: Roles of Arachidonic Acid and Its Metabolites in Pancreatic Physiology and Pathophysiology. Molecules 2023; 28:4316. [PMID: 37298790 PMCID: PMC10254454 DOI: 10.3390/molecules28114316] [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: 04/27/2023] [Revised: 05/17/2023] [Accepted: 05/19/2023] [Indexed: 06/12/2023] Open
Abstract
One of the most important constituents of the cell membrane is arachidonic acid. Lipids forming part of the cellular membrane can be metabolized in a variety of cellular types of the body by a family of enzymes termed phospholipases: phospholipase A2, phospholipase C and phospholipase D. Phospholipase A2 is considered the most important enzyme type for the release of arachidonic acid. The latter is subsequently subjected to metabolization via different enzymes. Three enzymatic pathways, involving the enzymes cyclooxygenase, lipoxygenase and cytochrome P450, transform the lipid derivative into several bioactive compounds. Arachidonic acid itself plays a role as an intracellular signaling molecule. Additionally, its derivatives play critical roles in cell physiology and, moreover, are involved in the development of disease. Its metabolites comprise, predominantly, prostaglandins, thromboxanes, leukotrienes and hydroxyeicosatetraenoic acids. Their involvement in cellular responses leading to inflammation and/or cancer development is subject to intense study. This manuscript reviews the findings on the involvement of the membrane lipid derivative arachidonic acid and its metabolites in the development of pancreatitis, diabetes and/or pancreatic cancer.
Collapse
Affiliation(s)
| | | | | | - Antonio González
- Instituto de Biomarcadores de Patologías Moleculares, Departamento de Fisiología, Universidad de Extremadura, 10003 Cáceres, Spain; (C.O.-P.); (A.C.-R.); (M.E.)
| |
Collapse
|
13
|
Zhang R, Kang R, Tang D. Ferroptosis in gastrointestinal cancer: From mechanisms to implications. Cancer Lett 2023; 561:216147. [PMID: 36965540 DOI: 10.1016/j.canlet.2023.216147] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 03/27/2023]
Abstract
Ferroptosis is a form of regulated cell death that is initiated by excessive lipid peroxidation that results in plasma membrane damage and the release of damage-associated molecular patterns. In recent years, ferroptosis has gained significant attention in cancer research due to its unique mechanism compared to other forms of regulated cell death, especially caspase-dependent apoptotic cell death. Gastrointestinal (GI) cancer encompasses malignancies that arise in the digestive tract, including the stomach, intestines, pancreas, colon, liver, rectum, anus, and biliary system. These cancers are a global health concern, with high incidence and mortality rates. Despite advances in medical treatments, drug resistance caused by defects in apoptotic pathways remains a persistent challenge in the management of GI cancer. Hence, exploring the role of ferroptosis in GI cancers may lead to more efficacious treatment strategies. In this review, we provide a comprehensive overview of the core mechanism of ferroptosis and discuss its function, regulation, and implications in the context of GI cancers.
Collapse
Affiliation(s)
- Ruoxi Zhang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
14
|
Paskaš S, Murganić B, Kuhnert R, Hey-Hawkins E, Mijatović S, Maksimović-Ivanić D. Carborane-Based Analog of Rev-5901 Attenuates Growth of Colon Carcinoma In Vivo. Molecules 2022; 27:molecules27144503. [PMID: 35889376 PMCID: PMC9321230 DOI: 10.3390/molecules27144503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/08/2022] [Accepted: 07/08/2022] [Indexed: 11/16/2022] Open
Abstract
Lipoxygenases convert polyunsaturated fatty acids into biologically active metabolites such as inflammatory mediators—prostaglandins and leukotrienes. The inhibition of lipoxygenases is increasingly employed in the treatment of cancer. We evaluated the anticancer potential of two novel 5-lipoxygenase inhibitors, named CarbZDNaph and CarbZDChin, which are analogues of the commercially available inhibitor Rev-5901. The in vitro segment of this study was conducted on a mouse colorectal carcinoma cell line—CT26CL25. For an in vivo model, we induced tumors in BALB/c mice by the implantation of CT26CL25 cells, and we treated the animals with potential inhibitors. A 48 h treatment resulted in diminished cell viability. Calculated IC50 values (half-maximal inhibitory concentrations) were 25 μM, 15 μM and 30 μM for CarbZDNaph, CarbZDChin and Rev-5901, respectively. The detailed analysis of mechanism revealed an induction of caspase-dependent apoptosis and autophagy. In the presence of chloroquine, an autophagy inhibitor, we observed an increased mortality of cells, implying a cytoprotective role of autophagy. Our in vivo experiment reports tumor growth attenuation in animals treated with CarbZDChin. Compounds CarbZDNaph and Rev-5901 lacked an in vivo efficacy. The results presented in this study display a strong effect of compound CarbZDChin on malignant cell growth. Having in mind the important role of inflammation in cancer development, these results have a significant impact and are worthy of further evaluation.
Collapse
Affiliation(s)
- Svetlana Paskaš
- Department of Immunology, Institute for Biological Research “SinišaStanković”, Belgrade University, 11060 Belgrade, Serbia; (S.P.); (B.M.); (S.M.)
| | - Blagoje Murganić
- Department of Immunology, Institute for Biological Research “SinišaStanković”, Belgrade University, 11060 Belgrade, Serbia; (S.P.); (B.M.); (S.M.)
| | - Robert Kuhnert
- Institute of Inorganic Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, 04103 Leipzig, Germany; (R.K.); (E.H.-H.)
| | - Evamarie Hey-Hawkins
- Institute of Inorganic Chemistry, Faculty of Chemistry and Mineralogy, Leipzig University, 04103 Leipzig, Germany; (R.K.); (E.H.-H.)
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research “SinišaStanković”, Belgrade University, 11060 Belgrade, Serbia; (S.P.); (B.M.); (S.M.)
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research “SinišaStanković”, Belgrade University, 11060 Belgrade, Serbia; (S.P.); (B.M.); (S.M.)
- Correspondence: ; Tel.: +381-11-2078452
| |
Collapse
|
15
|
Leukotrienes promote stem cell self-renewal and chemoresistance in acute myeloid leukemia. Leukemia 2022; 36:1575-1584. [PMID: 35461365 DOI: 10.1038/s41375-022-01579-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 03/30/2022] [Accepted: 04/13/2022] [Indexed: 11/09/2022]
Abstract
Acute myeloid leukemia (AML) is characterized by poor clinical outcomes due to high rates of relapse following standard-of-care induction chemotherapy. While many pathogenic drivers have been described in AML, our understanding of the molecular mechanisms mediating chemotherapy resistance remains poor. Therefore, we sought to identify resistance genes to induction therapy in AML and elucidated ALOX5 as a novel mediator of resistance to anthracycline-based therapy. ALOX5 is transcriptionally upregulated in AML patient blasts in comparison to normal hematopoietic stem/progenitor cells (HSPCs) and ALOX5 mRNA, and protein expression is increased in response to induction therapy. In vitro, and in vivo genetic, and pharmacologic perturbation studies confirm that ALOX5 positively regulates the leukemogenic potential of AML LSCs, and its loss does not significantly affect the function of normal HSPCs. ALOX5 mediates resistance to daunorubicin (DNR) and promotes AML cell survival and maintenance through its leukotriene (LT) synthetic capacity, specifically via modulating the synthesis of LTB4 and its binding to LTB receptor (BLTR). Our study reveals a previously unrecognized role of LTs in AML pathogenesis and chemoresistance, whereby inhibition of ALOX5 mediated LTB4 synthesis and function could be combined with standard chemotherapy, to enhance the overall therapeutic efficacy in AML.
Collapse
|
16
|
Blogowski W, Dolegowska K, Deskur A, Dolegowska B, Starzynska T. Lipoxins and Resolvins in Patients With Pancreatic Cancer: A Preliminary Report. Front Oncol 2022; 11:757073. [PMID: 35087747 PMCID: PMC8787076 DOI: 10.3389/fonc.2021.757073] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Eicosanoids are bioactive lipids derived from arachidonic acid, which have emerged as key regulators of a wide variety of pathophysiological processes in recent times and are implicated as mediators of gastrointestinal cancer. In this study, we investigated the systemic levels of lipoxygenase (LOX)-derived lipoxin A4 and B4, together with resolvin D1 and D2 in patients with pancreatic adenocarcinoma (n = 68), as well as in healthy individuals (n = 32). Systemic concentrations of the aforementioned immunoresolvents were measured using an enzyme-linked immunosorbent assay (ELISA). In this study, we observed that compared with concentrations in healthy individuals, the peripheral concentrations of the aforementioned eicosanoids were significantly elevated (2- to 10-fold) in patients with pancreatic cancer (in all cases p<0.00001). No significant association was observed between eicosanoid levels and the TNM clinical staging. Furthermore, we observed no significant differences in concentrations of the analyzed bioactive lipids between patients diagnosed with early-stage (TNM stage I-II) and more advanced disease (TNM stage III-IV). Receiver operating characteristic (ROC) curve analysis of each aforementioned immunoresolvent showed area under the curve values ranging between 0.79 and 1.00. Sensitivity, specificity, as well as positive and negative predictive values of the eicosanoids involved in the detection/differentiation of pancreatic adenocarcinoma ranged between 56.8% and 100%. In summary, our research is the first study that provides clinical evidence to support a systemic imbalance in LOX-derived lipoxins and resolvins as the mechanism underlying the pathogenesis of pancreatic adenocarcinoma. This phenomenon occurs regardless of the clinical TNM stage of the disease. Furthermore, our study is the first to preliminarily highlight the role of peripheral levels of immunoresolvents, particularly resolvin D1, as potential novel biomarkers of pancreatic cancer in humans.
Collapse
Affiliation(s)
- Wojciech Blogowski
- Institute of Medical Sciences, University of Zielona Gora, Zielona Gora, Poland
| | - Katarzyna Dolegowska
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Anna Deskur
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| | - Barbara Dolegowska
- Department of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Teresa Starzynska
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
17
|
Mai S, Inkielewicz-Stepniak I. Pancreatic Cancer and Platelets Crosstalk: A Potential Biomarker and Target. Front Cell Dev Biol 2021; 9:749689. [PMID: 34858977 PMCID: PMC8631477 DOI: 10.3389/fcell.2021.749689] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/04/2021] [Indexed: 12/12/2022] Open
Abstract
Platelets have been recognized as key players in hemostasis, thrombosis, and cancer. Preclinical and clinical researches evidenced that tumorigenesis and metastasis can be promoted by platelets through a wide variety of crosstalk between cancer cells and platelets. Pancreatic cancer is a devastating disease with high morbidity and mortality worldwide. Although the relationship between pancreatic cancer and platelets in clinical diagnosis is described, the interplay between pancreatic cancer and platelets, the underlying pathological mechanism and pathways remain a matter of intensive study. This review summaries recent researches in connections between platelets and pancreatic cancer. The existing data showed different underlying mechanisms were involved in their complex crosstalk. Typically, pancreatic tumor accelerates platelet aggregation which forms thrombosis. Furthermore, extracellular vesicles released by platelets promote communication in a neoplastic microenvironment and illustrate how these interactions drive disease progression. We also discuss the advantages of novel model organoids in pancreatic cancer research. A more in-depth understanding of tumor and platelets crosstalk which is based on organoids and translational therapies may provide potential diagnostic and therapeutic strategies for pancreatic cancer progression.
Collapse
Affiliation(s)
- Shaoshan Mai
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Iwona Inkielewicz-Stepniak
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
18
|
Li L, Xiao Y, Xu Z, Wang S. Zileuton inhibits arachidonate-5-lipoxygenase to exert antitumor effects in preclinical cervical cancer models. Cancer Chemother Pharmacol 2021; 88:953-960. [PMID: 34477945 DOI: 10.1007/s00280-021-04343-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/17/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND Inhibitors of arachidonate lipoxygenase 5 (ALOX5) exhibit anticancer activity. Zileuton is an FDA-approved drug for treating asthma and an ALOX5 inhibitor. This study evaluated the efficacy of zileuton in cervical cancer, determined the molecular mechanism of action, and assessed ALOX5 expression in cervical cancer patients. METHODS The effects of zileuton were evaluated using cervical cancer cell lines and xenograft mouse models. Loss-of-function analysis of ALOX5 was performed using siRNA. The levels of ALOX5 and 5-HETE were determined using immunohistochemistry and ELISA. RESULTS Zileuton resulted in cell proliferation inhibition and apoptosis induction in a dose-dependent manner, regardless of cellular origin or HPV infection. In two independent cervical cancer xenograft mouse models, zileuton at nontoxic doses significantly prevented tumor formation and decreased tumor growth. Zileuton acts on cervical cancer cells by inhibiting the ALOX5-5-HETE axis. Of note, ALOX5-5-HETE was significantly upregulated in cervical cancer compared with normal tissue. Inhibition of ALOX5 via the siRNA approach mimics the inhibitory effects of zileuton and confirms the roles of ALOX5 in cervical cancer. CONCLUSIONS Our work demonstrates that the ALOX5-5-HETE axis is activated in cervical cancer, with important roles in growth and survival, and this can be therapeutically targeted by zileuton. Our findings also provide preclinical evidence to assess the efficacy of zileuton in cervical cancer in clinical settings.
Collapse
Affiliation(s)
- Liling Li
- Department of Obstetrics and Gynecology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Yifang Xiao
- Department of Obstetrics and Gynecology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Zhengzheng Xu
- Department of Obstetrics and Gynecology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Shaoshuai Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jiefang Rd 1095, Qiaokou District, Wuhan, 430030, China.
| |
Collapse
|
19
|
Salgado MTSF, Lopes AC, Fernandes E Silva E, Cardoso JQ, Vidal RS, Cavalcante-Silva LHA, Carvalho DCM, Machado KDS, Rodrigues-Mascarenhas S, Rumjanek VM, Votto APDS. Relation between ABCB1 overexpression and COX2 and ALOX5 genes in human erythroleukemia cell lines. Prostaglandins Other Lipid Mediat 2021; 155:106553. [PMID: 33975019 DOI: 10.1016/j.prostaglandins.2021.106553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 04/23/2021] [Accepted: 05/05/2021] [Indexed: 10/21/2022]
Abstract
This study aimed to characterize the relationship between the COX2 and ALOX5 genes, as well as their link with the multidrug resistance (MDR) phenotype in sensitive (K562) and MDR (K562-Lucena and FEPS) erythroleukemia cells. For this, the inhibitors of 5-LOX (zileuton) and COX-2 (acetylsalicylic acid-ASA) and cells with the silenced ABCB1 gene were used. The treatment with ASA caused an increase in the gene expression of COX2 and ABCB1 in both MDR cell lines, and a decrease in the expression of ALOX5 in the FEPS cells. Silencing the ABCB1 gene induced a decrease in COX2 expression and an increase in the ALOX5 gene. Treatment with zileuton did not alter the expression of COX2 and ABCB1. Cytometry data showed that there was an increase in ABCB1 protein expression after exposure to ASA. In addition, the increased activity of ABCB1 in the K562-Lucena cell line indicates that ASA may be a substrate for this efflux pump, corroborating the molecular docking that showed that ASA can bind to ABCB1. Regardless of the genetic alteration in COX2 and ABCB1, the direct relationship between these genes and the inverse relationship with ALOX5 remained in the MDR cell lines. We assume that ABCB1 can play a regulatory role in COX2 and ALOX5 during the transformation of the parental cell line K562, explaining the increased gene expression of COX2 and decreased ALOX5 in the MDR cell lines.
Collapse
MESH Headings
- Humans
- Cyclooxygenase 2/genetics
- Cyclooxygenase 2/metabolism
- Arachidonate 5-Lipoxygenase/metabolism
- Arachidonate 5-Lipoxygenase/genetics
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- Leukemia, Erythroblastic, Acute/genetics
- Leukemia, Erythroblastic, Acute/pathology
- Leukemia, Erythroblastic, Acute/metabolism
- Hydroxyurea/pharmacology
- Hydroxyurea/analogs & derivatives
- Cell Line, Tumor
- K562 Cells
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
Collapse
Affiliation(s)
| | - Alessandra Costa Lopes
- Laboratório de Cultura Celular, ICB, FURG, RS, Brazil; Escola de Química e Alimentos, EQA, FURG, RS, Brazil
| | | | | | | | | | | | | | | | | | - Ana Paula de Souza Votto
- Laboratório de Cultura Celular, ICB, FURG, RS, Brazil; Programa de Pós-Graduação em Ciências Fisiológicas, ICB, FURG, RS, Brazil.
| |
Collapse
|
20
|
Tang J, Zhang C, Lin J, Duan P, Long J, Zhu H. ALOX5-5-HETE promotes gastric cancer growth and alleviates chemotherapy toxicity via MEK/ERK activation. Cancer Med 2021; 10:5246-5255. [PMID: 34121352 PMCID: PMC8335819 DOI: 10.1002/cam4.4066] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 04/16/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022] Open
Abstract
Background Recent studies highlight the regulatory role of arachidonate lipoxygenase5 (Alox5) and its metabolite 5‐hydroxyeicosatetraenoic acid (5‐HETE) in cancer tumorigenesis and progression. In this study, we analyzed the expression, biological function and the downstream signaling of Alox5 in gastric cancer. Methods Alox5 protein levels were measured using IHC and ELISA. Growth, migration and survival assays were performed. Phosphorylation of molecules involved in growth and survival signaling were analyzed by WB. Analysis of variance and t‐test were used for statistic analysis. Results Alox5 and 5‐HETE levels were upregulated in gastric cancer patients. ALOX5 overexpression or 5‐HETE addition activates gastric cancer cells and reduces chemotherapy’s efficacy. In contrast, ALOX5 inhibition via genetic and pharmacological approaches suppresses gastric cancer cells and enhances chemotherapy’s efficacy. In addition, Alox5 inhibition led to suppression of ERK‐mediated signaling pathways whereas ALOX5‐5‐HETE activates ERK‐mediated signaling in gastric cancer cells. Conclusions Our work demonstrates the critical role of ALOX5‐5‐HETE in gastric cancer and provides pre‐clinical evidence to initialize clinical trial using zileuton in combination with chemotherapy for treating gastric cancer.
Collapse
Affiliation(s)
- Jianjun Tang
- Department of General Surgery, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Chuang Zhang
- Department of Pediatrics, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Jingjing Lin
- Department of Blood Transfusion, Xiangyang Traditional Chinese Medicine Hospital, Xiangyang, China
| | - Peng Duan
- Department of Obstetrics and Gynaecology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Jian Long
- Department of Oncology, Jingzhou Central Hospital, The Second Clinical Medical College, Yangtze University, Jingzhou, China
| | - Hongyan Zhu
- Department of Oncology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| |
Collapse
|
21
|
Chen X, Kang R, Kroemer G, Tang D. Targeting ferroptosis in pancreatic cancer: a double-edged sword. Trends Cancer 2021; 7:891-901. [PMID: 34023326 DOI: 10.1016/j.trecan.2021.04.005] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/29/2021] [Accepted: 04/30/2021] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains an aggressive malignancy with a 5-year survival rate below 10%. Its unique genetic makeup and tumor microenvironment produce a lack of response to current treatments, including chemotherapy, radiotherapy, and immunotherapy. Recent preclinical studies have revealed that ferroptosis, an iron-dependent form of nonapoptotic cell death driven by unrestricted lipid peroxidation, may be an attractive therapeutic goal in PDAC. Understanding the dual role of ferroptotic cell death in both promoting and suppressing tumor immunity, as well as its integrated regulatory mechanisms and signaling pathways, may lead to more effective treatment designs for clinical trials of PDAC and may minimize or delay the emergence of drug resistance or side effects.
Collapse
Affiliation(s)
- Xin Chen
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, The Third Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China; Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Rui Kang
- Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe Labéllisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, 94800 Villejuif, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, Assistance Publique - Hôpitaux de Paris, 75015 Paris, France; Suzhou Institute for Systems Biology, Chinese Academy of Sciences, Suzhou, China; Department of Women's and Children's Health, Karolinska University Hospital, 17176 Stockholm, Sweden.
| | - Daolin Tang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, The Third Affiliated Hospital, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China; Department of Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
22
|
Mahgoub S, Hashad N, Ali S, Ibrahim R, Said AM, Moharram FA, Mady M. Polyphenolic Profile of Callistemon viminalis Aerial Parts: Antioxidant, Anticancer and In Silico 5-LOX Inhibitory Evaluations. Molecules 2021; 26:molecules26092481. [PMID: 33923148 PMCID: PMC8123052 DOI: 10.3390/molecules26092481] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/17/2021] [Accepted: 04/20/2021] [Indexed: 12/24/2022] Open
Abstract
Five new compounds viz kaempferol 3-O-(4″-galloyl)-β-d-glucopyranosyl-(1‴→6″)-O-β-d-glucopyranoside (1), kaempferol 3-O-β-d-mannuronopyranoside (2), kaempferol 3-O-β-d-mannopyranoside (3), quercetin 3-O-β-d-mannuronopyranoside (4), 2, 3 (S)- hexahydroxydiphenoyl]-d-glucose (5) along with fifteen known compounds were isolated from 80% aqueous methanol extract (AME) of C. viminalis. AME and compounds exerted similar or better antioxidant activity to ascorbic acid using DPPH, O2−, and NO inhibition methods. In addition, compounds 16, 4, and 7 showed cytotoxic activity against MCF-7 cell lines while 3, 7 and 16 exhibited strong activity against HepG2. An in silico analysis using molecular docking for polyphenolic compounds 2, 3, 7, 16 and 17 against human stable 5-LOX was performed and compared to that of ascorbic acid and quercetin. The binding mode as well as the enzyme-inhibitor interactions were evaluated. All compounds occupied the 5-LOX active site and showed binding affinity greater than ascorbic acid or quercetin. The data herein suggest that AME, a source of polyphenols, could be used against oxidative-stress-related disorders.
Collapse
Affiliation(s)
- Shahenda Mahgoub
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Ein-Helwan, Helwan, Cairo 11795, Egypt;
- Correspondence: (S.M.); (A.M.S.); (F.A.M.); Tel.: +1716-907-5016 (A.M.S.); +202-2554-160 (F.A.M.)
| | - Nashwa Hashad
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo 11795, Egypt; (N.H.); (R.I.); (M.M.)
| | - Sahar Ali
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Ein-Helwan, Helwan, Cairo 11795, Egypt;
| | - Reham Ibrahim
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo 11795, Egypt; (N.H.); (R.I.); (M.M.)
| | - Ahmed M. Said
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Helwan University, Ein-Helwan, Helwan, Cairo 11795, Egypt
- Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
- Correspondence: (S.M.); (A.M.S.); (F.A.M.); Tel.: +1716-907-5016 (A.M.S.); +202-2554-160 (F.A.M.)
| | - Fatma A. Moharram
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo 11795, Egypt; (N.H.); (R.I.); (M.M.)
- Correspondence: (S.M.); (A.M.S.); (F.A.M.); Tel.: +1716-907-5016 (A.M.S.); +202-2554-160 (F.A.M.)
| | - Mohamed Mady
- Department of Pharmacognosy, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo 11795, Egypt; (N.H.); (R.I.); (M.M.)
| |
Collapse
|
23
|
Muñoz-Osses M, Quiroz J, Vásquez-Martínez Y, Flores E, Navarrete E, Godoy F, Torrent C, Cortez-San Martín M, Gómez A, Mascayano C. Evaluation of cyrhetrenyl and ferrocenyl precursors as 5-lipoxygenase inhibitors – biological and computational studies. NEW J CHEM 2021. [DOI: 10.1039/d1nj01336j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Synthesis and biological evaluation of precursors derived from ferrocene and cyrhetrene as inhibitors of enzyme 5-hLOX.
Collapse
Affiliation(s)
| | - Javiera Quiroz
- Departamento Ciencias del Ambiente
- Universidad de Santiago de Chile
- Chile
| | - Yesseny Vásquez-Martínez
- Programa Centro de Investigaciones Biomédicas y Aplicadas (CIBAP)
- Escuela de Medicina
- Facultad de Ciencias Médicas
- Universidad de Santiago de Chile
- Chile
| | - Erick Flores
- Departamento Química de los Materiales
- Universidad de Santiago de Chile
- Chile
| | | | - Fernando Godoy
- Departamento Química de los Materiales
- Universidad de Santiago de Chile
- Chile
| | - Claudia Torrent
- Departamento Ciencias del Ambiente
- Universidad de Santiago de Chile
- Chile
| | | | - Alejandra Gómez
- Departamento Química de los Materiales
- Universidad de Santiago de Chile
- Chile
| | | |
Collapse
|
24
|
Cheng JH, Zhang WJ, Zhu JF, Cui D, Song KD, Qiang P, Mei CZ, Nie ZC, Ding BS, Han Z, Ding ZE, Zheng WW. CaMKIIγ regulates the viability and self-renewal of acute myeloid leukaemia stem-like cells by the Alox5/NF-κB pathway. Int J Lab Hematol 2020; 43:699-706. [PMID: 33369192 DOI: 10.1111/ijlh.13440] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/22/2020] [Accepted: 11/28/2020] [Indexed: 12/23/2022]
Abstract
Acute myeloid leukaemia (AML) is a frequently fatal malignant disease of haematopoietic stem and progenitor cells. The molecular and phenotypic characteristics of AML are highly heterogeneous. Our previous study concluded that CaMKIIγ was the trigger of chronic myeloid leukaemia progression from the chronic phase to blast crisis, but how CaMKIIγ influences AML stem-like cells remains elusive. In this study, we found that CaMKIIγ was overexpressed in AML patients and AML cell lines, as measured by qRT-PCR and Western blot assays. Moreover, CaMKIIγ decreased when the disease was in remission. Using an shRNA lentivirus expression system, we established CaMKIIγ stable-knockdown AML cell lines and found that knockdown of CaMKIIγ inhibited the viability and self-renewal of AML stem-like cell lines. Additionally, the ratio of CD34 + AML cell lines decreased, and CaMKIIγ knockdown induced the downregulation of Alox5 levels. We further detected downstream molecules of the Alox5/NF-κB pathway and found that c-myc and p-IκBα decreased while total IκBα remained normal. In conclusion, our study describes a new role for CaMKIIγ as a stem-like cell marker that is highly regulated by the Alox5/NF-κB pathway in AML stem-like cells. CaMKIIγ can participate in the viability and self-renewal of AML stem-like cells by regulating the Alox5/NF-κB pathway.
Collapse
Affiliation(s)
- Jiang-Hua Cheng
- School of Tea & Food Science, Anhui Agricultural University, Hefei, China.,Institute of Agro-products Processing Research, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Wen-Jing Zhang
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jun-Feng Zhu
- Department of Hematology, First Affiliated Hospital, Bengbu Medical College, Bengbu, China
| | - Di Cui
- School of Laboratory Medicine, Bengbu Medical College, Bengbu, China
| | - Kai-Di Song
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ping Qiang
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Chuan-Zhong Mei
- School of Laboratory Medicine, Bengbu Medical College, Bengbu, China
| | - Zheng-Chao Nie
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bang-Sheng Ding
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhong Han
- Department of Clinical Laboratory, Shengzhou People's Hospital, Shenzhou Branch of the First Affiliated Hospital of Zhejiang University, Shengzhou, China
| | - Zhi-En Ding
- School of Tea & Food Science, Anhui Agricultural University, Hefei, China.,Department of Biology and Food Engineering, Bozhou University, Bozhou, China
| | - Wei-Wei Zheng
- Department of Clinical Laboratory, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
25
|
Hsu KC, HuangFu WC, Lin TE, Chao MW, Sung TY, Chen YY, Pan SL, Lee JC, Tzou SC, Sun CM, Yang JM. A site-moiety map and virtual screening approach for discovery of novel 5-LOX inhibitors. Sci Rep 2020; 10:10510. [PMID: 32601404 PMCID: PMC7324578 DOI: 10.1038/s41598-020-67420-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 06/04/2020] [Indexed: 11/09/2022] Open
Abstract
The immune system works in conjunction with inflammation. Excessive inflammation underlies various human diseases, such as asthma, diabetes and heart disease. Previous studies found that 5-lipoxygenase (5-LOX) plays a crucial role in metabolizing arachidonic acid into inflammatory mediators and is a potential therapeutic target. In this study, we performed an in silico approach to establish a site-moiety map (SiMMap) to screen for new 5-LOX inhibitors. The map is composed of several anchors that contain key residues, moiety preferences, and their interaction types (i.e., electrostatic (E), hydrogen-bonding (H), and van der Waals (V) interactions) within the catalytic site. In total, we identified one EH, one H, and five V anchors, within the 5-LOX catalytic site. Based on the SiMMap, three 5-LOX inhibitors (YS1, YS2, and YS3) were identified. An enzyme-based assay validated inhibitory activity of YS1, YS2, and YS3 against 5-LOX with an IC50 value of 2.7, 4.2, and 5.3 μM, respectively. All three inhibitors significantly decrease LPS-induced TNF-α and IL-6 production, which suggests its potential use an anti-inflammatory agent. In addition, the identified 5-LOX inhibitors contain a novel scaffold. The discovery of these inhibitors presents an opportunity for designing specific anti-inflammatory drugs.
Collapse
Affiliation(s)
- Kai-Cheng Hsu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Biomedical Commercialization Center, Taipei Medical University, Taipei, Taiwan
| | - Wei-Chun HuangFu
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Tony Eight Lin
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Min-Wu Chao
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Ying Sung
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan
| | - Yi-Ying Chen
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Shiow-Lin Pan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
- Biomedical Commercialization Center, Taipei Medical University, Taipei, Taiwan
| | - Jih-Chin Lee
- Department of Otolaryngology-Head and Neck Surgery, Tri-Service General Hospital, Taipei, Taiwan
- Department of Otolaryngology-Head and Neck Surgery, National Defense Medical Center, Taipei, Taiwan
| | - Shey-Cherng Tzou
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan
| | - Chung-Ming Sun
- Department of Applied Chemistry, National Chiao Tung University, Hsinchu, Taiwan
| | - Jinn-Moon Yang
- Institute of Bioinformatics and Systems Biology, National Chiao Tung University, Hsinchu, Taiwan.
- Department of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.
- Center for Intelligent Drug Systems and Smart Bio-Devices, National Chiao Tung University, Hsinchu, Taiwan.
| |
Collapse
|
26
|
Gener P, Montero S, Xandri-Monje H, Díaz-Riascos ZV, Rafael D, Andrade F, Martínez-Trucharte F, González P, Seras-Franzoso J, Manzano A, Arango D, Sayós J, Abasolo I, Schwartz S. Zileuton™ loaded in polymer micelles effectively reduce breast cancer circulating tumor cells and intratumoral cancer stem cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 24:102106. [PMID: 31666201 DOI: 10.1016/j.nano.2019.102106] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/28/2019] [Accepted: 10/01/2019] [Indexed: 10/25/2022]
Abstract
Tumor recurrence, metastatic spread and progressive gain of chemo-resistance of advanced cancers are sustained by the presence of cancer stem cells (CSCs) within the tumor. Targeted therapies with the aim to eradicate these cells are thus highly regarded. However, often the use of new anti-cancer therapies is hampered by pharmacokinetic demands. Drug delivery through nanoparticles has great potential to increase efficacy and reduce toxicity and adverse effects. However, its production has to be based on intelligent design. Likewise, we developed polymeric nanoparticles loaded with Zileuton™, a potent inhibitor of cancer stem cells (CSCs), which was chosen based on high throughput screening. Its great potential for CSCs treatment was subsequently demonstrated in in vitro and in in vivo CSC fluorescent models. Encapsulated Zileuton™ reduces amount of CSCs within the tumor and effectively blocks the circulating tumor cells (CTCs) in the blood stream and metastatic spread.
Collapse
Affiliation(s)
- Petra Gener
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain.
| | - Sara Montero
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Helena Xandri-Monje
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Zamira V Díaz-Riascos
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Functional Validation & Preclinical Research (FVPR), CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Diana Rafael
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Fernanda Andrade
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, Universidade do Porto,Porto, Portugal
| | - Francesc Martínez-Trucharte
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Patricia González
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza, Spain
| | - Joaquin Seras-Franzoso
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Albert Manzano
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Diego Arango
- Biomedical Research in Digestive Tract Tumors, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joan Sayós
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza, Spain; Immune Regulation and Immunotherapy, CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ibane Abasolo
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Functional Validation & Preclinical Research (FVPR), CIBBIM-Nanomedicine, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza, Spain.
| | - Simo Schwartz
- Drug Delivery and Targeting Group, Molecular Biology and Biochemistry Research Centre for Nanomedicine (CIBBIM-Nanomedicine), Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain; Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Zaragoza, Spain.
| |
Collapse
|
27
|
Zileuton, a 5-Lipoxygenase Inhibitor, Exerts Anti-Angiogenic Effect by Inducing Apoptosis of HUVEC via BK Channel Activation. Cells 2019; 8:cells8101182. [PMID: 31575085 PMCID: PMC6829222 DOI: 10.3390/cells8101182] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 09/27/2019] [Accepted: 09/28/2019] [Indexed: 02/06/2023] Open
Abstract
The arachidonic acid metabolism through 5-lipoxygenase (5-LO) pathways is involved in modulating both tumorigenesis and angiogenesis. Although anti-carcinogenic activities of certain 5-LO inhibitors have been reported, the role of zileuton, a well known 5-LO inhibitor, on the endothelial cell proliferation and angiogenesis has not been fully elucidated. Here, we report that zileuton has an anti-angiogenic effect, and the underlying mechanisms involved activation of the large-conductance Ca2+-activated K+ (BK) channel. Our results show that zileuton significantly prevented vascular endothelial growth factor (VEGF)-induced proliferation of human umbilical vein endothelial cells (HUVECs) in vitro, as well as in vivo. However, such anti-angiogenic effect of zileuton was abolished by iberiotoxin (IBTX), a BK channel blocker, suggesting zileuton-induced activation of BK channel was critical for the observed anti-angiogenic effect of zileuton. Furthermore, the anti-angiogenic effect of zileuton was, at least, due to the activation of pro-apoptotic signaling cascades which was also abolished by IBTX. Additionally, zileuton suppressed the expression of VCAM-1, ICAM-1, ETS related gene (Erg) and the production of nitric oxide (NO). Taken together, our results show that zileuton prevents angiogenesis by activating the BK channel dependent-apoptotic pathway, thus highlighting its therapeutic capacity in angiogenesis-related diseases, such as cancer.
Collapse
|
28
|
Puratchikody A, Umamaheswari A, Irfan N, Sinha S, Manju SL, Ramanan M, Ramamoorthy G, Doble M. A novel class of tyrosine derivatives as dual 5-LOX and COX-2/mPGES1 inhibitors with PGE2 mediated anticancer properties. NEW J CHEM 2019. [DOI: 10.1039/c8nj04385j] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Leukotriene and prostaglandin pathways are controlled by the enzymes, LOX and COX/mPGES1 respectively and are responsible for inflammatory responses.
Collapse
Affiliation(s)
- Ayarivan Puratchikody
- Drug Discovery and Development Research Group
- Department of Pharmaceutical Technology
- Bharathidasan Institute of Technology
- Anna University
- Tiruchirappalli
| | - Appavoo Umamaheswari
- Drug Discovery and Development Research Group
- Department of Pharmaceutical Technology
- Bharathidasan Institute of Technology
- Anna University
- Tiruchirappalli
| | - Navabshan Irfan
- Drug Discovery and Development Research Group
- Department of Pharmaceutical Technology
- Bharathidasan Institute of Technology
- Anna University
- Tiruchirappalli
| | - Shweta Sinha
- Bioengineering and Drug Design Lab
- Department of Biotechnology
- Bhupat and Jyoti Mehta School of Biosciences
- Indian Institute of Technology
- Madras
| | - S. L. Manju
- Department of Chemistry
- Vellore Institute of Technology
- Vellore
- India
| | - Meera Ramanan
- Bioengineering and Drug Design Lab
- Department of Biotechnology
- Bhupat and Jyoti Mehta School of Biosciences
- Indian Institute of Technology
- Madras
| | - Gayathri Ramamoorthy
- Bioengineering and Drug Design Lab
- Department of Biotechnology
- Bhupat and Jyoti Mehta School of Biosciences
- Indian Institute of Technology
- Madras
| | - Mukesh Doble
- Bioengineering and Drug Design Lab
- Department of Biotechnology
- Bhupat and Jyoti Mehta School of Biosciences
- Indian Institute of Technology
- Madras
| |
Collapse
|
29
|
Khophai S, Thanee M, Techasen A, Namwat N, Klanrit P, Titapun A, Jarearnrat A, Sa-Ngiamwibool P, Loilome W. Zileuton suppresses cholangiocarcinoma cell proliferation and migration through inhibition of the Akt signaling pathway. Onco Targets Ther 2018; 11:7019-7029. [PMID: 30410359 PMCID: PMC6198876 DOI: 10.2147/ott.s178942] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Background Inflammatory lipid mediators play an important role in several cancer types. Leukotrienes (LTs), pro-inflammatory lipid mediators, are involved in chronic inflammation and cancer progression. They are derived from arachidonic acid by 5-lipoxygenase (5-LOX) activity. On the other hand, 15-lipoxygenase (15-LOX-1) converts LTs into lipoxins (LXs), pro-resolving lipid mediators. LXs are involved in the attenuation of inflammation and cancer development. Purpose We aimed to investigate the lipid mediator pathways, especially the LTs and LXs pathways, by studying 5-LOX and 15-LOX-1 expression in human cholangiocarcinoma (CCA) tissue. We also investigated the efficiency of zileuton (5-LOX inhibitor) treatment and BML-111 (LXA4 analog) addition on CCA cell lines properties. Patients and methods The expression of 5-LOX and 15-LOX-1 in fifty human cholangiocarcinoma (CCA) tissue was analyzed using immunohistochemical staining. In addition, the effect of zileuton and BML-111 on CCA cell growth and migration was demonstrated using a cell viability assay and wound-healing assay, respectively. Furthermore, the molecular mechanism by which zileuton inhibits CCA cell migration was revealed using immunofluorescent staining and western blot analysis, respectively. Results We demonstrate that the upregulation of 5-LOX is significantly correlated with CCA recurrent status. A positive 15-LOX-1 signal was significantly associated with a longer survival time in CCA patients. We found that co-expression of 5-LOX and 15-LOX-1 resulted in a relatively good prognosis in CCA patients. In addition, zileuton could inhibit CCA cell migration as well as BML-111. Interestingly, zileuton treatment not only downregulated 5-LOX, but also upregulated 15-LOX-1, together with reversing the epithelial-mesenchymal transition to mesenchymal-epithelial transition phenotype as observed in EMT marker western blot. Conclusion These findings suggest that 5-LOX and 15-LOX-1 play a key role in CCA and may serve as targets for CCA therapy.
Collapse
Affiliation(s)
- Sasikamon Khophai
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand, .,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand,
| | - Malinee Thanee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand, .,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand,
| | - Anchalee Techasen
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand, .,Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Nisana Namwat
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand, .,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand,
| | - Poramate Klanrit
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand, .,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand,
| | - Attapol Titapun
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand, .,Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Apiwat Jarearnrat
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand, .,Department of Surgery, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Prakasit Sa-Ngiamwibool
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand, .,Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Watcharin Loilome
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand, .,Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand,
| |
Collapse
|
30
|
Merchant N, Bhaskar LVKS, Momin S, Sujatha P, Reddy ABM, Nagaraju GP. 5-Lipoxygenase: Its involvement in gastrointestinal malignancies. Crit Rev Oncol Hematol 2018; 127:50-55. [PMID: 29891111 DOI: 10.1016/j.critrevonc.2018.05.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 04/16/2018] [Accepted: 05/14/2018] [Indexed: 12/11/2022] Open
Abstract
Lipoxygenases (LOXs) are dioxygenases that catalyze the peroxidation of linoleic acid (LA) or arachidonic acid (AA), in the presence of molecular oxygen. The existence of inflammatory component in the tumor microenvironment intimately links the LOXs to gastrointestinal (GI) cancer progression. Amongst the six-different human LOX-isoforms, 5-LOX is the most vital enzyme for leukotriene (LT) biosynthesis, which is the main inflammation intermediaries. As recent investigations have shown the association of 5-LOX with tumor metastasis, there has also been significant progress in discovering the function of 5-LOX pathway in GI cancer. Studies on GI cancer cells using the pharmacological drugs targeting 5-LOX pathway have shown antiproliferative and proapoptotic effects. Pharmacogenetic discoveries in other diseases have revealed strong heritable basis for the leukotriene pathway, which helps in exploring the mechanistic source of genetic alteration within the leukotriene pathway and offer insights into GI cancer pathogenesis and future prospects for treatment and prevention. This review recapitulates the current research status of 5-LOX activity in GI malignancies.
Collapse
Affiliation(s)
- Neha Merchant
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | | | - Saimila Momin
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA
| | - Peela Sujatha
- Department of Biotechnology, Dr. B.R. Ambedkar University, Etcherla, Srikakulam, Andhra Pradesh, 532410, India
| | - Aramati B M Reddy
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
31
|
Jain R, Austin Pickens C, Fenton JI. The role of the lipidome in obesity-mediated colon cancer risk. J Nutr Biochem 2018; 59:1-9. [PMID: 29605789 DOI: 10.1016/j.jnutbio.2018.02.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 02/07/2018] [Accepted: 02/13/2018] [Indexed: 02/07/2023]
Abstract
Obesity is a state of chronic inflammation influenced by lipids such as fatty acids and their secondary oxygenated metabolites deemed oxylipids. Many such lipid mediators serve as potent signaling molecules of inflammation, which can further alter lipid metabolism and lead to carcinogenesis. For example, sphingosine-1-phosphate activates cyclooxygenase-2 in endothelial cells resulting in the conversion of arachidonic acid (AA) to prostaglandin E2 (PGE2). PGE2 promotes colon cancer cell growth. In contrast, the less studied path of AA oxygenation via cytochrome p450 enzymes produces epoxyeicosatetraenoic acids (EETs), whose anti-inflammatory properties cause shrinking of enlarged adipocytes, a characteristic of obesity, through the liberation of fatty acids. It is now thought that EET depletion occurs in obesity and may contribute to colon cell carcinogenesis. Meanwhile, gangliosides, a type of sphingolipid, are cell surface signaling molecules that contribute to the apoptosis of colon tumor cells. Many of these discoveries have been made recently and the mechanisms are still not fully understood, leading to an exciting new chapter of lipidomic research. In this review, mechanisms behind obesity-associated colon cancer are discussed with a focus on the role of small lipid signaling molecules in the process. Specifically, changes in lipid metabolite levels during obesity and the development of colon cancer, as well as novel biomarkers and targets for therapy, are discussed.
Collapse
Affiliation(s)
- Raghav Jain
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA
| | - C Austin Pickens
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA
| | - Jenifer I Fenton
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
32
|
Piromkraipak P, Sangpairoj K, Tirakotai W, Chaithirayanon K, Unchern S, Supavilai P, Power C, Vivithanaporn P. Cysteinyl Leukotriene Receptor Antagonists Inhibit Migration, Invasion, and Expression of MMP-2/9 in Human Glioblastoma. Cell Mol Neurobiol 2018; 38:559-573. [PMID: 28600709 PMCID: PMC11481984 DOI: 10.1007/s10571-017-0507-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/06/2017] [Indexed: 12/21/2022]
Abstract
Glioblastoma is one of the most malignant and aggressive types of brain tumors. 5-lipoxygenase and cysteinyl leukotriene receptor 1 (CysLT1) play a role in human carcinogenesis. Leukotriene receptor antagonists (LTRAs), anti-asthmatic drugs with mild side effects, have anti-metastatic activity in epidermoid carcinoma, lung carcinoma, and colon cancers as well as neuroprotective effects. Herein, anti-migratory effects of two LTRAs, montelukast and zafirlukast, were investigated in glioblastoma cells. The level of CysLT1 in A172 cells was increased by 3.13 folds after IL-1β treatment. The median toxic concentration of LTRAs in A172, U373, and primary astrocytes ranged from 7.17 to 26.28 μM at 24-h post-exposure. Both LTRAs inhibited migration and invasion of glioma. Additionally, both drugs significantly inhibited the expression and activities of MMP-2 and MMP-9 in A172 and U373 glioblastoma cells and primary human astrocytes, suggesting that CysLT1 plays a role in migration and invasion of glioma, and LTRAs are potential drugs to reduce migration and invasion.
Collapse
Affiliation(s)
- Pannaree Piromkraipak
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Kant Sangpairoj
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok, Thailand
- Division of Anatomy, Department of Preclinical Science, Faculty of Medicine, Thammasat University, Pathum Thani, Thailand
| | | | | | - Supeenun Unchern
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Porntip Supavilai
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Christopher Power
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Pornpun Vivithanaporn
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
33
|
Liao D, Qian B, Zhang Y, Wu K, Xu M. Inhibition of 5-lipoxygenase represses neutrophils activation and activates apoptosis in pancreatic tissues during acute necrotizing pancreatitis. Biochem Biophys Res Commun 2018; 498:79-85. [PMID: 29421656 DOI: 10.1016/j.bbrc.2018.02.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 02/03/2018] [Indexed: 11/29/2022]
Abstract
Pancreatic glandular necrosis is rapid inflammation of the pancreas and contributes to severe acute pancreatitis in humans. The pathogenesis of pancreatic tissue inflammation during acute pancreatitis is still largely unknown. Recent studies suggest that 5-lipoxygenase (5-LOX) is an essential mediator in modulating cell death pathways in human diseases. In this study, we aimed to evaluate the effects of a 5-LOX inhibitor, zileuton, on tissue apoptosis and neutrophils activation in pancreatic tissues during acute necrotizing pancreatitis (ANP) in a rat model. In this present study, both mRNA and protein levels of 5-LOX are upregulated during ANP and zileuton treatment is shown to repress ANP-induced upregulation of 5-LOX levels. In addition, zileuton treatment is found to repress blood biomarkers of neutrophils activation such as soluble intercellular adhesive molecular 1 (ICAM-1), soluble E-selectin (E-selectin), soluble P-selectin (P-selectin), leukotriene B4 (LTB4), and myeloperoxidase (MPO). Also, zileuton treatment attenuates pancreatic tissue pathology, upregulates caspase-3, downregulates B-cell lymphoma 2 (Bcl-2), and activates tissue apoptosis evaluated by TUNEL staining. Our results show that 5-LOX plays an important role in activating apoptosis and repressing neutrophils activation during ANP. The current study suggests that 5-LOX can be used as a potential target for the treatment of ANP.
Collapse
Affiliation(s)
- Dan Liao
- Department of Gastroenterology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Bo Qian
- Department of Gastroenterology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Yefei Zhang
- Department of Gastroenterology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Kai Wu
- Department of Gastroenterology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Min Xu
- Department of Gastroenterology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, 200080, China.
| |
Collapse
|
34
|
Rezende BM, Athayde RM, Gonçalves WA, Resende CB, Teles de Tolêdo Bernardes P, Perez DA, Esper L, Reis AC, Rachid MA, Castor MGME, Cunha TM, Machado FS, Teixeira MM, Pinho V. Inhibition of 5-lipoxygenase alleviates graft-versus-host disease. J Exp Med 2017; 214:3399-3415. [PMID: 28947611 PMCID: PMC5679175 DOI: 10.1084/jem.20170261] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Revised: 07/24/2017] [Accepted: 08/18/2017] [Indexed: 01/26/2023] Open
Abstract
Rezende et al. report that the transplant of 5-lipoxygenase (5-LO)−deficient leukocytes protects mice from GVHD. Treatment with the 5-LO inhibitor zileuton or a LTB4 antagonist at the initial phase of the transplant achieves similar protective effects. 5-LO is a crucial contributor to tissue damage in GVHD. Leukotriene B4 (LTB4), a proinflammatory mediator produced by the enzyme 5-lipoxygenase (5-LO), is associated with the development of many inflammatory diseases. In this study, we evaluated the participation of the 5-LO/LTB4 axis in graft-versus-host disease (GVHD) pathogenesis by transplanting 5-LO–deficient leukocytes and investigated the effect of pharmacologic 5-LO inhibition by zileuton and LTB4 inhibition by CP-105,696. Mice that received allogeneic transplant showed an increase in nuclear 5-LO expression in splenocytes, indicating enzyme activation after GVHD. Mice receiving 5-LO–deficient cell transplant or zileuton treatment had prolonged survival, reduced GVHD clinical scores, reduced intestinal and liver injury, and decreased levels of serum and hepatic LTB4. These results were associated with inhibition of leukocyte recruitment and decreased production of cytokines and chemokines. Treatment with CP-105,696 achieved similar effects. The chimerism or the beneficial graft-versus-leukemia response remained unaffected. Our data provide evidence that the 5-LO/LTB4 axis orchestrates GVHD development and suggest it could be a target for the development of novel therapeutic strategies for GVHD treatment.
Collapse
Affiliation(s)
- Barbara Maximino Rezende
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Rayssa Maciel Athayde
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - William Antônio Gonçalves
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Carolina Braga Resende
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Priscila Teles de Tolêdo Bernardes
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Denise Alves Perez
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Lísia Esper
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Alesandra Côrte Reis
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Milene Alvarenga Rachid
- Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Marina Gomes Miranda E Castor
- Departamento de Farmacologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Thiago Mattar Cunha
- Departamento de Farmacologia, Faculdade de Medicina, Universidade de São Paulo, Ribeirão Preto, São Paulo, Brasil
| | - Fabiana Simão Machado
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Mauro Martins Teixeira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| | - Vanessa Pinho
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brasil
| |
Collapse
|
35
|
ALOX5 exhibits anti-tumor and drug-sensitizing effects in MLL-rearranged leukemia. Sci Rep 2017; 7:1853. [PMID: 28500307 PMCID: PMC5431828 DOI: 10.1038/s41598-017-01913-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 03/30/2017] [Indexed: 12/30/2022] Open
Abstract
MLL-rearranged acute myeloid leukemia (AML) remains a fatal disease with a high rate of relapse and therapeutic failure due to chemotherapy resistance. In analysis of our Affymetrix microarray profiling and chromatin immunoprecipitation (ChIP) assays, we found that ALOX5 is especially down-regulated in MLL-rearranged AML, via transcription repression mediated by Polycomb repressive complex 2 (PRC2). Colony forming/replating and bone marrow transplantation (BMT) assays showed that Alox5 exhibited a moderate anti-tumor effect both in vitro and in vivo. Strikingly, leukemic cells with Alox5 overexpression showed a significantly higher sensitivity to the standard chemotherapeutic agents, i.e., doxorubicin (DOX) and cytarabine (Ara-C). The drug-sensitizing role of Alox5 was further confirmed in human and murine MLL-rearranged AML cell models in vitro, as well as in the in vivo MLL-rearranged AML BMT model coupled with treatment of “5 + 3” (i.e. DOX plus Ara-C) regimen. Stat and K-Ras signaling pathways were negatively correlated with Alox5 overexpression in MLL-AF9-leukemic blast cells; inhibition of the above signaling pathways mimicked the drug-sensitizing effect of ALOX5 in AML cells. Collectively, our work shows that ALOX5 plays a moderate anti-tumor role and functions as a drug sensitizer, with a therapeutic potential, in MLL-rearranged AML.
Collapse
|
36
|
Bynigeri RR, Jakkampudi A, Jangala R, Subramanyam C, Sasikala M, Rao GV, Reddy DN, Talukdar R. Pancreatic stellate cell: Pandora's box for pancreatic disease biology. World J Gastroenterol 2017; 23:382-405. [PMID: 28210075 PMCID: PMC5291844 DOI: 10.3748/wjg.v23.i3.382] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 11/09/2016] [Accepted: 12/19/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic stellate cells (PSCs) were identified in the early 1980s, but received much attention after 1998 when the methods to isolate and culture them from murine and human sources were developed. PSCs contribute to a small proportion of all pancreatic cells under physiological condition, but are essential for maintaining the normal pancreatic architecture. Quiescent PSCs are characterized by the presence of vitamin A laden lipid droplets. Upon PSC activation, these perinuclear lipid droplets disappear from the cytosol, attain a myofibroblast like phenotype and expresses the activation marker, alpha smooth muscle actin. PSCs maintain their activated phenotype via an autocrine loop involving different cytokines and contribute to progressive fibrosis in chronic pancreatitis (CP) and pancreatic ductal adenocarcinoma (PDAC). Several pathways (e.g., JAK-STAT, Smad, Wnt signaling, Hedgehog etc.), transcription factors and miRNAs have been implicated in the inflammatory and profibrogenic function of PSCs. The role of PSCs goes much beyond fibrosis/desmoplasia in PDAC. It is now shown that PSCs are involved in significant crosstalk between the pancreatic cancer cells and the cancer stroma. These interactions result in tumour progression, metastasis, tumour hypoxia, immune evasion and drug resistance. This is the rationale for therapeutic preclinical and clinical trials that have targeted PSCs and the cancer stroma.
Collapse
|
37
|
Microarray-based gene expression profiling reveals genes and pathways involved in the oncogenic function of REG3A on pancreatic cancer cells. Gene 2016; 578:263-73. [DOI: 10.1016/j.gene.2015.12.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Revised: 10/18/2015] [Accepted: 12/16/2015] [Indexed: 01/05/2023]
|