51
|
Doxorubicin Paradoxically Ameliorates Tumor-Induced Inflammation in Young Mice. Int J Mol Sci 2021; 22:ijms22169023. [PMID: 34445729 PMCID: PMC8396671 DOI: 10.3390/ijms22169023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/29/2021] [Accepted: 08/17/2021] [Indexed: 01/03/2023] Open
Abstract
Doxorubicin (DOX) is one of the most widely used chemo-therapeutic agents in pediatric oncology. DOX elicits an inflammatory response in multiple organs, which contributes to DOX-induced adverse effects. Cancer itself causes inflammation leading to multiple pathologic conditions. The current study investigated the inflammatory response to DOX and tumors using an EL4-lymphoma, immunocompetent, juvenile mouse model. Four-week old male C57BL/6N mice were injected subcutaneously with EL4 lymphoma cells (5 × 104 cells/mouse) in the flank region, while tumor-free mice were injected with vehicle. Three days following tumor implantation, both tumor-free and tumor-bearing mice were injected intraperitoneally with either DOX (4 mg/kg/week) or saline for 3 weeks. One week after the last DOX injection, the mice were euthanized and the hearts, livers, kidneys, and serum were harvested. Gene expression and serum concentration of inflammatory markers were quantified using real-time PCR and ELISA, respectively. DOX treatment significantly suppressed tumor growth in tumor-bearing mice and caused significant cardiac atrophy in tumor-free and tumor-bearing mice. EL4 tumors elicited a strong inflammatory response in the heart, liver, and kidney. Strikingly, DOX treatment ameliorated tumor-induced inflammation paradoxical to the effect of DOX in tumor-free mice, demonstrating a widely divergent effect of DOX treatment in tumor-free versus tumor-bearing mice.
Collapse
|
52
|
Chemotherapy: a double-edged sword in cancer treatment. Cancer Immunol Immunother 2021; 71:507-526. [PMID: 34355266 DOI: 10.1007/s00262-021-03013-3] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/07/2021] [Indexed: 12/24/2022]
Abstract
Chemotherapy is a well-known and effective treatment for different cancers; unfortunately, it has not been as efficient in the eradication of all cancer cells as been expected. The mechanism of this failure was not fully clarified, yet. Meanwhile, alterations in the physiologic conditions of the tumor microenvironment (TME) were suggested as one of the underlying possibilities. Chemotherapy drugs can activate multiple signaling pathways and augment the secretion of inflammatory mediators. Inflammation may show two opposite roles in the TME. On the one hand, inflammation, as an innate immune response, tries to suppress tumor growth but on the other hand, it might be not powerful enough to eradicate the cancer cells and even it can provide appropriate conditions for cancer promotion and relapse as well. Therefore, the administration of mild anti-inflammatory drugs during chemotherapy might result in more successful clinical results. Here, we will review and discuss this hypothesis. Most chemotherapy agents are triggers of inflammation in the tumor microenvironment through inducing the production of senescence-associated secretory phenotype (SASP) molecules. Some chemotherapy agents can induce systematic inflammation by provoking TLR4 signaling or triggering IL-1B secretion through the inflammasome pathway. NF-kB and MAPK are key signaling pathways of inflammation and could be activated by several chemotherapy drugs. Furthermore, inflammation can play a key role in cancer development, metastasis and exacerbation.
Collapse
|
53
|
Quagliariello V, De Laurentiis M, Rea D, Barbieri A, Monti MG, Carbone A, Paccone A, Altucci L, Conte M, Canale ML, Botti G, Maurea N. The SGLT-2 inhibitor empagliflozin improves myocardial strain, reduces cardiac fibrosis and pro-inflammatory cytokines in non-diabetic mice treated with doxorubicin. Cardiovasc Diabetol 2021; 20:150. [PMID: 34301253 PMCID: PMC8305868 DOI: 10.1186/s12933-021-01346-y] [Citation(s) in RCA: 242] [Impact Index Per Article: 60.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 07/16/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Empagliflozin (EMPA), a selective inhibitor of the sodium glucose co-transporter 2, reduced the risk of hospitalization for heart failure and cardiovascular death in type 2 diabetic patients in the EMPA-REG OUTCOME trial. Recent trials evidenced several cardio-renal benefits of EMPA in non-diabetic patients through the involvement of biochemical pathways that are still to be deeply analysed. We aimed to evaluate the effects of EMPA on myocardial strain of non-diabetic mice treated with doxorubicin (DOXO) through the analysis of NLRP3 inflammasome and MyD88-related pathways resulting in anti-apoptotic and anti-fibrotic effects. METHODS Preliminary cellular studies were performed on mouse cardiomyocytes (HL-1 cell line) exposed to doxorubicin alone or combined to EMPA. The following analysis were performed: determination of cell viability (through a modified MTT assay), study of intracellular ROS production, lipid peroxidation (quantifying intracellular malondialdehyde and 4-hydroxynonenal), intracellular Ca2+ homeostasis. Moreover, pro-inflammatory studies were also performed: expression of NLRP3 inflammasome, MyD88 myddosome and p65/NF-κB associated to secretion of cytokines involved in cardiotoxicity (Interleukins 1β, 8, 6). C57Bl/6 mice were untreated (Sham, n = 6) or treated for 10 days with doxorubicin (DOXO, n = 6), EMPA (EMPA, n = 6) or doxorubicin combined to EMPA (DOXO-EMPA, n = 6). DOXO was injected intraperitoneally. Ferroptosis and xanthine oxidase were studied before and after treatments. Cardiac function studies, including EF, FS and radial/longitudinal strain were analysed through transthoracic echocardiography (Vevo 2100). Cardiac fibrosis and apoptosis were histologically studied through Picrosirius red and TUNEL assay, respectively and quantified through pro-collagen-1α1, MMP-9 and Caspase-3 expression. Tissue NLRP3, MyD88 and cytokines were also quantified before and after treatments through ELISA methods. RESULTS Cardiomyocytes exposed to doxorubicin increased the intracellular Ca2+ content and expression of several pro-inflammatory markers associated to cell death; co-incubation with EMPA reduced significantly the magnitude of the effects. In preclinical study, EMPA increased EF and FS compared to DOXO groups (p < 0.05), prevented the reduction of radial and longitudinal strain after 10 days of treatment with doxorubicin (RS) 30.3% in EMPA-DOXO vs 15.7% in DOXO mice; LS - 17% in EMPA-DOXO vs - 11.7% in DOXO mice (p < 0.001 for both). Significant reductions in ferroptosis, xanthine oxidase expression, cardiac fibrosis and apoptosis in EMPA associated to DOXO were also seen. A reduced expression of pro-inflammatory cytokines, NLRP3, MyD88 and NF-kB in heart, liver and kidneys was also seen in DOXO-EMPA group compared to DOXO (p < 0.001). CONCLUSION EMPA reduced ferroptosis, fibrosis, apoptosis and inflammation in doxorubicin-treated mice through the involvement of NLRP3 and MyD88-related pathways, resulting in significant improvements in cardiac functions. These findings provides the proof of concept for translational studies designed to reduce adverse cardiovascular outcomes in non-diabetic cancer patients treated with doxorubicin.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy.
| | | | - Domenica Rea
- SSD Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Antonio Barbieri
- SSD Sperimentazione Animale, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Maria Gaia Monti
- Department of Translational Medical Sciences, University of Naples "Federico II", Naples, Italy
| | - Andreina Carbone
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Andrea Paccone
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Via L. De Crecchio 7, 80138, Naples, Italy
| | - Mariarosaria Conte
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Via L. De Crecchio 7, 80138, Naples, Italy
| | - Maria Laura Canale
- Cardiology Division, Azienda USL Toscana Nord-Ovest, Versilia Hospital, Lido Di Camaiore, Italy
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy.
| |
Collapse
|
54
|
Liu Q, Hua M, Zhang C, Wang R, Liu J, Yang X, Han F, Hou M, Ma D. NLRP3-activated bone marrow dendritic cells play antileukemic roles via IL-1β/Th1/IFN-γ in acute myeloid leukemia. Cancer Lett 2021; 520:109-120. [PMID: 34237408 DOI: 10.1016/j.canlet.2021.06.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/01/2021] [Accepted: 06/15/2021] [Indexed: 01/09/2023]
Abstract
The bone marrow microenvironment of acute myeloid leukemia (AML) characterized by immunosuppressive features fosters leukemia immune escape. Elucidating the immunosuppressive mechanism and developing effective immunotherapeutic strategies are necessary. Here, we found that the Th1% and IFN-γ level were downregulated in bone marrow of AML and NLRP3-activated BMDCs promoted CD4+ T cell differentiation into Th1 cells via IL-1β secretion. However, IFN-γ-producing Th1 cells were not induced by NLRP3-activated BMDCs in the presence of the NLRP3 inflammasome inhibitor MCC950 or anti-IL-1β antibody in vitro unless exogenous IL-1β was replenished. This inhibitory effect on Th1 differentiation was also observed in Nlrp3-/- mice or anti-IL-1β antibody-treated mice. Notably, elevated Th1 cell levels promoted apoptosis and inhibited proliferation in leukemia cells via IFN-γ secretion in vitro and in vivo. Thus, NLRP3-activated BMDCs promote the proliferation of IFN-γ-producing Th1 cells with antileukemic effects and may provide insight into the basis for leukemia immunotherapy in patients with AML.
Collapse
Affiliation(s)
- Qinqin Liu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China; Department of Hematology, Taian Central Hospital, Taian, Shandong, 271000, China
| | - Mingqiang Hua
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Chen Zhang
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China; Department of Hematology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, China
| | - Ruiqing Wang
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Jinting Liu
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Xinyu Yang
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Fengjiao Han
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China
| | - Daoxin Ma
- Department of Hematology, Qilu Hospital, Shandong University, Jinan, 250012, China.
| |
Collapse
|
55
|
Fabiani I, Aimo A, Grigoratos C, Castiglione V, Gentile F, Saccaro LF, Arzilli C, Cardinale D, Passino C, Emdin M. Oxidative stress and inflammation: determinants of anthracycline cardiotoxicity and possible therapeutic targets. Heart Fail Rev 2021; 26:881-890. [PMID: 33319255 PMCID: PMC8149360 DOI: 10.1007/s10741-020-10063-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/01/2020] [Indexed: 12/04/2022]
Abstract
Chemotherapy with anthracycline-based regimens remains a cornerstone of treatment of many solid and blood tumors but is associated with a significant risk of cardiotoxicity, which can manifest as asymptomatic left ventricular dysfunction or overt heart failure. These effects are typically dose-dependent and cumulative and may require appropriate screening strategies and cardioprotective therapies in order to minimize changes to anticancer regimens or even their discontinuation. Our current understanding of cardiac damage by anthracyclines includes a central role of oxidative stress and inflammation. The identification of these processes through circulating biomarkers or imaging techniques might then be helpful for early diagnosis and risk stratification. Furthermore, therapeutic strategies relieving oxidative stress and inflammation hold promise to prevent heart failure development or at least to mitigate cardiac damage, although further evidence is needed on their efficacy, either alone or as part of combination therapies with neurohormonal antagonists, which are the current adopted standard.
Collapse
Affiliation(s)
- Iacopo Fabiani
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
| | - Alberto Aimo
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy.
| | | | | | | | - Luigi F Saccaro
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | | | - Daniela Cardinale
- Cardioncology Unit, European Institute of Oncology, IRCCS, Milan, Italy
| | - Claudio Passino
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Michele Emdin
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Pisa, Italy
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| |
Collapse
|
56
|
Gong H, Yuan N, Shen Z, Tang C, Shipp S, Qian L, Lu Y, Andolina IM, Zhang S, Wu J, Yang H, Wang W. Transduction catalysis: Doxorubicin amplifies rAAV-mediated gene expression in the cortex of higher-order vertebrates. iScience 2021; 24:102685. [PMID: 34195565 PMCID: PMC8233199 DOI: 10.1016/j.isci.2021.102685] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/24/2021] [Accepted: 06/01/2021] [Indexed: 12/26/2022] Open
Abstract
Rapid and efficient gene transduction via recombinant adeno-associated viruses (rAAVs) is highly desirable across many basic and clinical research domains. Here, we report that vector co-infusion with doxorubicin, a clinical anti-cancer drug, markedly enhanced rAAV-mediated transgene expression in the cerebral cortex across mammalian species (cat, mouse, and macaque), acting throughout the time period examined and detectable at just three days after transfection. This enhancement showed serotype generality, being common to all rAAV serotypes tested (2, 8, 9, and PHP.eB) and was observed both locally and at remote locations consistent with doxorubicin undergoing retrograde axonal transport. All these effects were observed at doses matching human blood plasma levels in clinical therapy and lacked detectable cytotoxicity as assessed by cell morphology, activity, apoptosis, and behavioral testing. Altogether, this study identifies an effective means to improve the capability and scope of in vivo rAAV applications, amplifying cell transduction at doxorubicin concentrations paralleling medical practice.
Collapse
Affiliation(s)
- Hongliang Gong
- Institute of Neuroscience, the Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Nini Yuan
- Institute of Neuroscience, the Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Zhiming Shen
- Institute of Neuroscience, the Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Cheng Tang
- Institute of Neuroscience, the Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Stewart Shipp
- Institute of Neuroscience, the Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Liling Qian
- Institute of Neuroscience, the Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Yiliang Lu
- Institute of Neuroscience, the Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ian Max Andolina
- Institute of Neuroscience, the Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shenghai Zhang
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Shanghai 200031, China
| | - Jihong Wu
- Department of Ophthalmology, Eye and ENT Hospital of Fudan University, Shanghai 200031, China
| | - Hui Yang
- Institute of Neuroscience, the Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Shanghai Center for Brain and Brain-Inspired Intelligence Technology, Shanghai 200031, China
| | - Wei Wang
- Institute of Neuroscience, the Center for Excellence in Brain Science and Intelligence Technology, State Key Laboratory of Neuroscience, Key Laboratory of Primate Neurobiology, Chinese Academy of Sciences, Shanghai 200031, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,Shanghai Center for Brain and Brain-Inspired Intelligence Technology, Shanghai 200031, China
| |
Collapse
|
57
|
ERDEM GÜZEL E. Pleurotus Eryngii Ekstraktının Sprague-Dawley Sıçanlarında Adriamisin Kaynaklı Kardiyotoksisite Üzerindeki Etkilerinin İncelenmesi. DICLE MEDICAL JOURNAL 2021. [DOI: 10.5798/dicletip.944395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
58
|
Maayah ZH, Alam AS, Takahara S, Soni S, Ferdaoussi M, Matsumura N, Zordoky BN, Eisenstat DD, Dyck JRB. Resveratrol reduces cardiac NLRP3-inflammasome activation and systemic inflammation to lessen doxorubicin-induced cardiotoxicity in juvenile mice. FEBS Lett 2021; 595:1681-1695. [PMID: 33876420 DOI: 10.1002/1873-3468.14091] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/22/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022]
Abstract
Doxorubicin (DOX) is a very effective anticancer agent that is widely used in pediatric cancer patients. Nevertheless, DOX is known to have cardiotoxic effects that may progress to cardiomyopathy later in life. We have recently shown that cotreatment of resveratrol (RES) with DOX in juvenile mice attenuates late-onset hypertension-induced cardiomyopathy. However, the molecular mechanism responsible for these changes remains unknown. Herein, we show that the cardiac NLRP3 inflammasome plays a crucial role in regulating cardiac injury in a DOX -treated juvenile mouse model and the detrimental effects of hypertension in these mice later in life. We further demonstrate that RES significantly reduces systemic inflammation to contribute to the improvements observed in DOX -induced cardiac injury in young mice and late-onset hypertension-induced cardiomyopathy.
Collapse
Affiliation(s)
- Zaid H Maayah
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Abrar S Alam
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Shingo Takahara
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Shubham Soni
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mourad Ferdaoussi
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Nobutoshi Matsumura
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Beshay N Zordoky
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN, USA
| | - David D Eisenstat
- Departments of Oncology, Medical Genetics and Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Jason R B Dyck
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
59
|
Shaldoum F, El-Kott AF, Ouda MMA, Abd-Ella EM. Immunomodulatory effects of bee pollen on doxorubicin-induced bone marrow/spleen immunosuppression in rat. J Food Biochem 2021; 45:e13747. [PMID: 33949702 DOI: 10.1111/jfbc.13747] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/22/2021] [Accepted: 04/15/2021] [Indexed: 01/24/2023]
Abstract
This study investigated the immunomodulatory effects of Bee Pollen (BP) on Doxorubicin (DOX)-induced bone marrow/spleen suppression in rats. 48 Wistar rats were divided into 6 groups (n = 8/group); control, DOX (5 mg/kg), BP (100 mg/kg), BP (200 mg/kg), BP (100 mg/kg) +DOX, and BP (200 mg/kg) +DOX groups. BP was administered orally for 42 days and 5 mg/kg of DOX was injected intravenously at days 7, 14, 21, 28, 35 and 42. Hematological parameters, antioxidant enzymes and inflammatory cytokines were measured. Apoptosis-related genes were investigated using Real-Time PCR and western blot. DOX significantly decreased blood cells count, cytokines, and antioxidant enzyme. It also increased the expression of apoptotic genes in spleen and BM. The BP significantly improved hematopoietic function, antioxidant parameters, and serum levels of hematopoietic simulating-cytokines. Also, BP significantly reduced the expression of apoptotic genes. These results confirm the immunomodulatory activity of BP in DOX-induced biochemical, molecular and histological immunosuppression. PRACTICAL APPLICATIONS: Chemotherapy drugs are being developed every day but are limited due to their side effects. The most important side effect of chemotherapy drugs is the suppression of hematopoiesis through its direct effect on bone marrow and hematopoietic cells. Today, many studies are done on natural, synthetic and semi-synthetic compounds to reduce the effects of chemotherapy drugs. Compounds that, along with chemotherapy drugs in the treatment of various tumors, maintain the hematopoietic pathway, synergize the antitumor effects of chemotherapy drugs, and also protect other organs of the body from free radical damage produced by chemotherapy drugs. One of these natural compounds is bee pollen, which has all the properties mentioned in chemotherapy supplements and can be used in the pharmaceutical industry.
Collapse
Affiliation(s)
- Fayez Shaldoum
- Department of Biology, College of Science, Jouf University, Sakaka, Saudi Arabia.,Department of Zoology, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Attalla F El-Kott
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia.,Zoology Department, College of Science, Damanhour University, Damanhour, Egypt
| | - Marwa Mohamed Ahmed Ouda
- Department of Nursing, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia.,Nursing Department, Faculty of Nursing, Damanhour University, Damanhour, Egypt
| | - Eman M Abd-Ella
- Zoology Department, College of Science, Fayoum University, Fayoum, Egypt.,Biology Department, College of Science and Art, Al-Baha University, Al-Mandaq, Saudi Arabia
| |
Collapse
|
60
|
Altinoz E, Oner Z, Elbe H, Uremis N, Uremis M. Linalool exhibits therapeutic and protective effects in a rat model of doxorubicin-induced kidney injury by modulating oxidative stress. Drug Chem Toxicol 2021; 45:2024-2030. [PMID: 33682561 DOI: 10.1080/01480545.2021.1894751] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
The aim of the present study was to investigate the therapeutic and protective effects of linalool against doxorubicin (DOX)-induced kidney injury. Forty-eight Wistar rats were divided into 8 groups as follows; Control, DOX [20 mg/kg, intraperitoneal (ip) single dose DOX], linalool (LIN50 and LIN100; 50 mg/kg and 100 mg/kg linalool via ip for 5 days, respectively), DOX + LIN50 and DOX + LIN100 (20 mg/kg single dose of DOX via ip on first day and 50 mg/kg and 100 mg/kg linalool via ip, respectively), LIN50 + DOX and LIN100 + DOX (50 mg/kg and 100 mg/kg linalool via ip for 5 days, respectively and 20 mg/kg single dose of DOX via ip on fifth day). Doxorubicin led to a significant increase in the level of malondialdehyde (MDA) in the kidney, whereas superoxide dismutase (SOD), catalase (CAT) and reduced glutathione (GSH) levels decreased remarkably when compared with control. On the other hand, LIN supplementation before and after DOX treatment led to a significant decrease in MDA and also increases in SOD, CAT and GSH levels. DOX caused significant increases in the levels of blood urea nitrogen (BUN) and creatinine (Cr) levels in the plasma, while LIN supplementation as a therapeutic and preventive agent led to significant decreases in BUN and Cr levels. The current study demonstrated that LIN supplementation after or before DOX treatment can led to therapeutic and preventive effects against DOX-induced renal damage.
Collapse
Affiliation(s)
- Eyup Altinoz
- Department of Medical Biochemistry, Faculty of Medicine, Karabuk University, Karabuk, Turkey
| | - Zulal Oner
- Department of Anatomy, Faculty of Medicine, Karabuk University, Karabuk, Turkey
| | - Hulya Elbe
- Department of Histology and Embryology, Faculty of Medicine, Mugla Sitki Kocman University, Mugla, Turkey
| | - Nuray Uremis
- Department of Medical Biochemistry, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Muhammed Uremis
- Department of Medical Biochemistry, Faculty of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
61
|
Alves MT, Simões R, Pestana RMC, de Oliveira AN, Oliveira HHM, Soares CE, Sabino ADP, Silva LM, Gomes KB. Interleukin-10 Levels are Associated with Doxorubicin-Related Cardiotoxicity in Breast Cancer Patients in a One-Year Follow-Up Study. Immunol Invest 2021; 51:883-898. [PMID: 33557640 DOI: 10.1080/08820139.2021.1882486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: Myocardial toxicity is a common side effect of doxorubicin (DOXO) therapy in breast cancer patients. We hypothesized that DOXO-induced cardiotoxicity may be related to the release of inflammatory cytokines in response to the treatment. This study aimed to assess changes in plasma levels of interleukin (IL)-1β, IL-6, IL-10 and tumor necrosis factor (TNF) after chemotherapy and to correlate these levels with cardiac biomarkers and clinical data.Methods: Sixty-four patients with breast cancer treated with DOXO were included. Twenty-two subjects (cases) developed cardiotoxicity until one year after the end of DOXO treatment. Cytokines and cardiac markers were evaluated before starting chemotherapy (T0), up to 7 days after the last infusion (T1) and 12 months after the last infusion (T2).Results: Higher IL-10 levels were observed in the case group compared to controls at T1 (p = .006) and T2 (p = .046). The IL-1β, IL-6 and TNF levels did not change during treatment in each group (p > .05), nor between the case and control groups. The IL-10 levels were higher at T1 than at T0 and T2 (p < .05 for both) in the cardiotoxicity group. A correlation between IL-10 and N-terminal pro-brain natriuretic peptide (NT-proBNP) levels at T0 and T2 in the cardiotoxicity group was observed (p = .048 and p = .004, respectively).Conclusion: Our study demonstrated that DOXO induced an increase in plasma IL-10 levels in patients who presented cardiotoxicity after treatment, which correlated with NT-proBNP levels.
Collapse
Affiliation(s)
- Michelle Teodoro Alves
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo Simões
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Faculdade Ciências Médicas de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo Mendonça Cardoso Pestana
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - Cintia Esteves Soares
- Fundação Hospitalar do Estado de Minas Gerais (FHEMIG), Belo Horizonte, Minas Gerais, Brazil
| | - Adriano de Paula Sabino
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Karina Braga Gomes
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
62
|
Cheong A, McGrath S, Robinson T, Maliki R, Spurling A, Lock P, Rephaeli A, Nudelman A, Parker BS, Pepe S, Cutts SM. A switch in mechanism of action prevents doxorubicin-mediated cardiac damage. Biochem Pharmacol 2021; 185:114410. [PMID: 33428897 DOI: 10.1016/j.bcp.2021.114410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 01/03/2021] [Accepted: 01/05/2021] [Indexed: 10/22/2022]
Abstract
Cancer patients treated with doxorubicin are at risk of congestive heart failure due to doxorubicin-mediated cardiotoxicity via topoisomerase IIβ poisoning. Acute cardiac muscle damage occurs in response to the very first dose of doxorubicin, however, cardioprotection has been reported after co-treatment of doxorubicin with acyloxyalkyl ester prodrugs. The aim of this study was to examine the role played by various forms of acute cardiac damage mediated by doxorubicin and determine a mechanism for the cardioprotective effect of formaldehyde-releasing prodrug AN-9 (pivaloyloxymethyl butyrate). Doxorubicin-induced cardiac damage in BALB/c mice bearing mammary tumours was established with a single dose of doxorubicin (4 or 16 mg/kg) administered alone or in combination with AN-9 (100 mg/kg). AN-9 protected the heart from doxorubicin-induced myocardial apoptosis and also significantly reduced dsDNA breaks, independent from the level of doxorubicin biodistribution to the heart. Covalent incorporation of [14C]doxorubicin into DNA showed that the combination treatment yielded significantly higher levels of formaldehyde-mediated doxorubicin-DNA adducts compared to doxorubicin alone, yet this form of damage was associated with cardioprotection from apoptosis. The cardiac transcriptomic analysis indicates that the combination treatment initiates inflammatory response signalling pathways. Doxorubicin and AN-9 combination treatments were cardioprotective, yet preserved doxorubicin-mediated anti-tumour proliferation and apoptosis in mammary tumours. This was associated with a switch in doxorubicin action from cardiac topoisomerase IIβ poisoning to covalent-DNA adduct formation. Co-administration of doxorubicin and formaldehyde-releasing prodrugs, such as AN-9, may be a promising cardioprotective therapy while maintaining doxorubicin activity in primary mammary tumours.
Collapse
Affiliation(s)
- Alison Cheong
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Sean McGrath
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Tina Robinson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Ruqaya Maliki
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Alex Spurling
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Peter Lock
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Ada Rephaeli
- Laboratory for Pharmacology and Experimental Oncology, Felsenstein Medical Research Center, Petach Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, 49100 Tel Aviv, Israel
| | - Abraham Nudelman
- Division of Medicinal Chemistry, Department of Chemistry, Bar-Ilan University, Ramat Gan 52900, Israel
| | - Belinda S Parker
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Salvatore Pepe
- Murdoch Children's Research Institute, Department of Cardiology, Royal Children's Hospital, Australia; Department of Paediatrics, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Suzanne M Cutts
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia.
| |
Collapse
|
63
|
Gefitinib initiates sterile inflammation by promoting IL-1β and HMGB1 release via two distinct mechanisms. Cell Death Dis 2021; 12:49. [PMID: 33414419 PMCID: PMC7791030 DOI: 10.1038/s41419-020-03335-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 12/09/2020] [Accepted: 12/11/2020] [Indexed: 12/20/2022]
Abstract
Anticancer drug gefitinib causes inflammation-based side effects, such as interstitial pneumonitis. However, its mechanisms remain unknown. Here, we provide evidence that gefitinib elicits pro-inflammatory responses by promoting mature-interleukin-1β (IL-1β) and high-mobility group box 1 (HMGB1) release. Mitochondrial reactive oxygen species (mtROS) driven by gefitinib stimulated the formation of the NLRP3 (NACHT, LRR and PYD-containing protein 3) inflammasome, leading to mature-IL-1β release. Notably, gefitinib also stimulated HMGB1 release, which is, however, not mediated by the NLRP3 inflammasome. On the other hand, gefitinib-driven mtROS promoted the accumulation of γH2AX, a hallmark of DNA damage, leading to the activation of poly (ADP-ribose) polymerase-1 (PARP-1) and subsequent active release of HMGB1. Together our results reveal the potential ability of gefitinib to initiate sterile inflammation via two distinct mechanisms, and identified IL-1β and HMGB1 as key determinants of gefitinib-induced inflammation that may provide insights into gefitinib-induced interstitial pneumonitis.
Collapse
|
64
|
Heinze-Milne SD, Keats MR, Blanchard C, Giacomantonio N, MacDonald D, Rajda M, Younis T, Grandy SA. Exercise to Prevent Anthracycline-Based Cardiotoxicity (EXACT): A Feasibility Study. TRANSLATIONAL JOURNAL OF THE AMERICAN COLLEGE OF SPORTS MEDICINE 2021. [DOI: 10.1249/tjx.0000000000000168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
65
|
Aljobaily N, Viereckl MJ, Hydock DS, Aljobaily H, Wu TY, Busekrus R, Jones B, Alberson J, Han Y. Creatine Alleviates Doxorubicin-Induced Liver Damage by Inhibiting Liver Fibrosis, Inflammation, Oxidative Stress, and Cellular Senescence. Nutrients 2020; 13:nu13010041. [PMID: 33374297 PMCID: PMC7824063 DOI: 10.3390/nu13010041] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/11/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022] Open
Abstract
Background: Treatment with the chemotherapy drug doxorubicin (DOX) may lead to toxicities that affect non-cancer cells including the liver. Supplementing the diet with creatine (Cr) has been suggested as a potential intervention to minimize DOX-induced side effects, but its effect in alleviating DOX-induced hepatoxicity is currently unknown. Therefore, we aimed to examine the effects of Cr supplementation on DOX-induced liver damage. Methods: Male Sprague-Dawley rats were fed a diet supplemented with 2% Cr for four weeks, 4% Cr for one week followed by 2% Cr for three more weeks, or control diet for four weeks. Animals then received either a bolus i.p. injection of DOX (15 mg/kg) or saline as a placebo. Animals were then sacrificed five days-post injection and markers of hepatoxicity were analyzed using the liver-to-body weight ratio, aspartate transaminase (AST)-to- alanine aminotransferase (ALT) ratio, alkaline phosphatase (ALP), lipemia, and T-Bilirubin. In addition, hematoxylin and eosin (H&E) staining, Picro-Sirius Red staining, and immunofluorescence staining for CD45, 8-OHdG, and β-galactosidase were performed to evaluate liver morphology, fibrosis, inflammation, oxidative stress, and cellular senescence, respectively. The mRNA levels for biomarkers of liver fibrosis, inflammation, oxidative stress, and senescence-related genes were measured in liver tissues. Chromosomal stability was evaluated using global DNA methylation ELISA. Results: The ALT/AST ratio and liver to body weight ratio tended to increase in the DOX group, and Cr supplementation tended to attenuate this increase. Furthermore, elevated levels of liver fibrosis, inflammation, oxidative stress, and senescence were observed with DOX treatment, and Cr supplementation prior to DOX treatment ameliorated this hepatoxicity. Moreover, DOX treatment resulted in chromosomal instability (i.e., altered DNA methylation profile), and Cr supplementation showed a tendency to restore chromosomal stability with DOX treatment. Conclusion: The data suggest that Cr protected against DOX-induced hepatotoxicity by attenuating fibrosis, inflammation, oxidative stress, and senescence.
Collapse
Affiliation(s)
- Nouf Aljobaily
- School of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA; (N.A.); (M.J.V.); (J.A.)
| | - Michael J. Viereckl
- School of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA; (N.A.); (M.J.V.); (J.A.)
| | - David S. Hydock
- School of Sport and Exercise Science, University of Northern Colorado, Greeley, CO 80639, USA; (D.S.H.); (R.B.); (B.J.)
| | | | - Tsung-Yen Wu
- Obstetrics and Gynecology Department, University of Washington, Seattle, WA 98115, USA;
| | - Raquel Busekrus
- School of Sport and Exercise Science, University of Northern Colorado, Greeley, CO 80639, USA; (D.S.H.); (R.B.); (B.J.)
| | - Brandon Jones
- School of Sport and Exercise Science, University of Northern Colorado, Greeley, CO 80639, USA; (D.S.H.); (R.B.); (B.J.)
| | - Jammie Alberson
- School of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA; (N.A.); (M.J.V.); (J.A.)
| | - Yuyan Han
- School of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA; (N.A.); (M.J.V.); (J.A.)
- Correspondence: ; Tel.: +1-970-351-2004
| |
Collapse
|
66
|
Transferrin-Bound Doxorubicin Enhances Apoptosis and DNA Damage through the Generation of Pro-Inflammatory Responses in Human Leukemia Cells. Int J Mol Sci 2020; 21:ijms21249390. [PMID: 33321722 PMCID: PMC7764702 DOI: 10.3390/ijms21249390] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022] Open
Abstract
Doxorubicin (DOX) is an effective antineoplastic drug against many solid tumors and hematological malignancies. However, the clinical use of DOX is limited, because of its unspecific mode of action. Since leukemia cells overexpress transferrin (Tf) receptors on their surface, we proposed doxorubicin–transferrin (DOX–Tf) conjugate as a new vehicle to increase drug concentration directly in cancer cells. The data obtained after experiments performed on K562 and CCRF-CEM human leukemia cell lines clearly indicate severe cytotoxic and genotoxic properties of the conjugate drug. On the other hand, normal peripheral blood mononuclear cells (PBMCs) were more resistant to DOX–Tf than to DOX. In comparison to free drug, we observed that Tf-bound DOX induced apoptosis in a TRAIL-dependent manner and caused DNA damage typical of programmed cell death. These fatal hallmarks of cell death were confirmed upon morphological observation of cells incubated with DOX or DOX–Tf. Studies of expression of TNF-α, IL-4, and IL-6 at the mRNA and protein levels revealed that the pro-inflammatory response plays an important role in the toxicity of the conjugate. Altogether, the results demonstrated here describe a mechanism of the antitumor activity of the DOX–Tf conjugate.
Collapse
|
67
|
Oxidative Stress-Inducing Anticancer Therapies: Taking a Closer Look at Their Immunomodulating Effects. Antioxidants (Basel) 2020; 9:antiox9121188. [PMID: 33260826 PMCID: PMC7759788 DOI: 10.3390/antiox9121188] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/19/2020] [Accepted: 11/25/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer cells are characterized by higher levels of reactive oxygen species (ROS) compared to normal cells as a result of an imbalance between oxidants and antioxidants. However, cancer cells maintain their redox balance due to their high antioxidant capacity. Recently, a high level of oxidative stress is considered a novel target for anticancer therapy. This can be induced by increasing exogenous ROS and/or inhibiting the endogenous protective antioxidant system. Additionally, the immune system has been shown to be a significant ally in the fight against cancer. Since ROS levels are important to modulate the antitumor immune response, it is essential to consider the effects of oxidative stress-inducing treatments on this response. In this review, we provide an overview of the mechanistic cellular responses of cancer cells towards exogenous and endogenous ROS-inducing treatments, as well as the indirect and direct antitumoral immune effects, which can be both immunostimulatory and/or immunosuppressive. For future perspectives, there is a clear need for comprehensive investigations of different oxidative stress-inducing treatment strategies and their specific immunomodulating effects, since the effects cannot be generalized over different treatment modalities. It is essential to elucidate all these underlying immune effects to make oxidative stress-inducing treatments effective anticancer therapy.
Collapse
|
68
|
Exercise Reduces the Resumption of Tumor Growth and Proteolytic Pathways in the Skeletal Muscle of Mice Following Chemotherapy. Cancers (Basel) 2020; 12:cancers12113466. [PMID: 33233839 PMCID: PMC7699885 DOI: 10.3390/cancers12113466] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Doxorubicin is a chemotherapeutic agent that contributes to muscle wasting. Based on the evidence that many cancer variants are associated with cachexia and that cancer patients are usually treated with chemotherapeutic agents, it is important to determine strategies to mitigate muscle atrophy. Muscle loss is a poor prognosis during cancer treatment, and exercise has emerged as a potential strategy utilized in this context. Once an ongoing regimen of chemotherapeutic treatment is not always possible, our results demonstrated that continuity of endurance exercise is a potential strategy that can be adopted when chemotherapy needs to be interrupted, minimizing the resumption of tumor growth and avoiding muscle loss. Abstract The pathogenesis of muscle atrophy plays a central role in cancer cachexia, and chemotherapy contributes to this condition. Therefore, the present study aimed to evaluate the effects of endurance exercise on time-dependent muscle atrophy caused by doxorubicin. For this, C57 BL/6 mice were subcutaneously inoculated with Lewis lung carcinoma cells (LLC group). One week after the tumor establishment, a group of these animals initiated the doxorubicin chemotherapy alone (LLC + DOX group) or combined with endurance exercise (LLC + DOX + EXER group). One group of animals was euthanized after the chemotherapy cycle, whereas the remaining animals were euthanized one week after the last administration of doxorubicin. The practice of exercise combined with chemotherapy showed beneficial effects such as a decrease in tumor growth rate after chemotherapy interruption and amelioration of premature death due to doxorubicin toxicity. Moreover, the protein degradation levels in mice undergoing exercise returned to basal levels after chemotherapy; in contrast, the mice treated with doxorubicin alone experienced an increase in the mRNA expression levels of the proteolytic pathways in gastrocnemius muscle (Trim63, Fbxo32, Myostatin, FoxO). Collectively, our results suggest that endurance exercise could be utilized during and after chemotherapy for mitigating muscle atrophy promoted by doxorubicin and avoid the resumption of tumor growth.
Collapse
|
69
|
Mancilla TR, Davis LR, Aune GJ. Doxorubicin-induced p53 interferes with mitophagy in cardiac fibroblasts. PLoS One 2020; 15:e0238856. [PMID: 32960902 PMCID: PMC7508395 DOI: 10.1371/journal.pone.0238856] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
Anthracyclines are the critical component in a majority of pediatric chemotherapy regimens due to their broad anticancer efficacy. Unfortunately, the vast majority of long-term childhood cancer survivors will develop a chronic health condition caused by their successful treatments and severe cardiac disease is a common life-threatening outcome that is unequivocally linked to previous anthracycline exposure. The intricacies of how anthracyclines such as doxorubicin, damage the heart and initiate a disease process that progresses over multiple decades is not fully understood. One area left largely unstudied is the role of the cardiac fibroblast, a key cell type in cardiac maturation and injury response. In this study, we demonstrate the effect of doxorubicin on cardiac fibroblast function in the presence and absence of the critical DNA damage response protein p53. In wildtype cardiac fibroblasts, doxorubicin-induced damage correlated with decreased proliferation and migration, cell cycle arrest, and a dilated cardiomyopathy gene expression profile. Interestingly, these doxorubicin-induced changes were completely or partially restored in p53-/- cardiac fibroblasts. Moreover, in wildtype cardiac fibroblasts, doxorubicin produced DNA damage and mitochondrial dysfunction, both of which are well-characterized cell stress responses induced by cytotoxic chemotherapy and varied forms of heart injury. A 3-fold increase in p53 (p = 0.004) prevented the completion of mitophagy (p = 0.032) through sequestration of Parkin. Interactions between p53 and Parkin increased in doxorubicin-treated cardiac fibroblasts (p = 0.0003). Finally, Parkin was unable to localize to the mitochondria in wildtype cardiac fibroblasts, but mitochondrial localization was restored in p53-/- cardiac fibroblasts. These findings strongly suggest that cardiac fibroblasts are an important myocardial cell type that merits further study in the context of doxorubicin treatment. A more robust knowledge of the role cardiac fibroblasts play in the development of doxorubicin-induced cardiotoxicity will lead to novel clinical strategies that will improve the quality of life of cancer survivors.
Collapse
Affiliation(s)
- T. R. Mancilla
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center San Antonio, San Antonio, TX, United States of America
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center San Antonio, San Antonio, TX, United States of America
| | - L. R. Davis
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center San Antonio, San Antonio, TX, United States of America
| | - G. J. Aune
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center San Antonio, San Antonio, TX, United States of America
- Department of Pediatrics, Division of Hematology-Oncology, University of Texas Health Science Center San Antonio, San Antonio, TX, United States of America
| |
Collapse
|
70
|
Hurmach Y, Rudyk M, Prylutska S, Hurmach V, Prylutskyy YI, Ritter U, Scharff P, Skivka L. C 60 Fullerene Governs Doxorubicin Effect on Metabolic Profile of Rat Microglial Cells In Vitro. Mol Pharm 2020; 17:3622-3632. [PMID: 32673486 DOI: 10.1021/acs.molpharmaceut.0c00691] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background: C60 fullerenes and their derivatives are actively investigated for the use in neuroscience. Applications of these nanoscale materials require the examination of their interaction with different neural cells, especially with microglia, because these cells, like other tissue resident phagocytes, are the earliest and most sensitive responders to nanoparticles. The aim of this study was to investigate the effect of C60 fullerene and its nanocomplex with doxorubicin (Dox) on the metabolic profile of brain-resident phagocytes-microglia-in vitro. Methods: Resting microglial cells from adult male Wistar rats were used in experiments. Potential C60 fullerene targets in microglial cells were studied by computer simulation. Microglia oxidative metabolism and phagocytic activity were examined by flow cytometry. Griess reaction and arginase activity colorimetric assay were used to explore arginine metabolism. Results: C60 fullerene when used alone did not influence microglia oxidative metabolism and phagocytic activity but shifted arginine metabolism toward the decrease of NO generation. Complexation of C60 fullerene with Dox (C60-Dox) potentiated the ability of the latter to stimulate NO generation. Conclusion: The capability of C60 fullerenes used alone to cause anti-inflammatory shift of microglia arginine metabolism makes them a promising agent for the correction of neuroinflammatory processes involved in neurodegeneration. The potentiating action of C60 fullerene on the immunomodulatory effect of Dox allows us to consider the C60 molecule as an attractive vehicle for this antitumor agent.
Collapse
Affiliation(s)
- Yevheniia Hurmach
- Bogomolets National Medical University, 13, T. Shevchenko blvd, 01601 Kyiv, Ukraine
| | - Mariia Rudyk
- Taras Shevchenko National University of Kyiv, Volodymyrska str., 64, 01601 Kyiv, Ukraine
| | - Svitlana Prylutska
- Taras Shevchenko National University of Kyiv, Volodymyrska str., 64, 01601 Kyiv, Ukraine
| | - Vasyl Hurmach
- Taras Shevchenko National University of Kyiv, Volodymyrska str., 64, 01601 Kyiv, Ukraine
| | - Yuriy I Prylutskyy
- Taras Shevchenko National University of Kyiv, Volodymyrska str., 64, 01601 Kyiv, Ukraine
| | - Uwe Ritter
- Institute of Chemistry and Biotechnology, Technical University of Ilmenau, Weimarer Str., 25, 98693 Ilmenau, Germany
| | - Peter Scharff
- Institute of Chemistry and Biotechnology, Technical University of Ilmenau, Weimarer Str., 25, 98693 Ilmenau, Germany
| | - Larysa Skivka
- Taras Shevchenko National University of Kyiv, Volodymyrska str., 64, 01601 Kyiv, Ukraine
| |
Collapse
|
71
|
Hashemzaei M, Mamoulakis C, Tsarouhas K, Georgiadis G, Lazopoulos G, Tsatsakis A, Shojaei Asrami E, Rezaee R. Crocin: A fighter against inflammation and pain. Food Chem Toxicol 2020; 143:111521. [DOI: 10.1016/j.fct.2020.111521] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/07/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023]
|
72
|
Li H, Xia B, Chen W, Zhang Y, Gao X, Chinnathambi A, Alharbi SA, Zhao Y. Nimbolide prevents myocardial damage by regulating cardiac biomarkers, antioxidant level, and apoptosis signaling against doxorubicin-induced cardiotoxicity in rats. J Biochem Mol Toxicol 2020; 34:e22543. [PMID: 32627270 DOI: 10.1002/jbt.22543] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 05/08/2020] [Accepted: 05/29/2020] [Indexed: 12/31/2022]
Abstract
The current work planned to assess the protecting properties of nimbolide against doxorubicin (DOX)-treated myocardial damage. Myocardial damage was produced with 2.5 mg/kg of DOX given on alternative days (14 days). Thiobarbituric acid reactive substances (TBARS) levels of a lipid peroxidative marker were elevated, whereas reduced body weight, heart weight, blood pressure indices and reduced levels of antioxidants like glutathione-S-transferase, superoxide dismutase, catalase, glutathione peroxidase, glutathione, and glutathione reductase were observed in the heart tissue of DOX-treated animals. DOX-treated animals showed augmented levels of cardiac markers likes monocyte chemotactic protein-1, interferon-gamma, aspartate transferase, creatine kinase, lactate dehydrogenase, creatine kinase-muscle/brain, heart-type fatty acid-binding protein, glycogen phosphorylase isoenzyme BB, transforming growth factor-β, brain natriuretic peptide, myoglobin, and cTnI in serum. Histopathological assessment confirmed the DOX-induced cardiotoxicity. Furthermore, DOX-induced rats showed augmented inflammatory mediators (nuclear factor-κB [NF-kB], tumor necrosis factor-α [TNF-α], and interleukin-1β [IL-1β]) and increased PI3K/Akt signaling proteins (PI3K, p-Bad/Bad, caspase-3, and p-Akt), whereas decreased oxidative markers (HO-1 and NQO-1) and p-PTEN were observed. Nimbolide-supplemented rats showed reduced activity/levels of cardiac markers and TBARS levels in serum and heart tissue. Levels of enzymatic and nonenzymatic antioxidants were augmented in the heart tissue of nimbolide-supplemented rats. Nimbolide influence decreased apoptosis, inflammation, and enhanced antioxidant markers through the modulation of p-Bad/Bad, caspase-3, PI3K, p-Akt, TNF-α, NF-kB, IL-1β, HO-1, NQO-1, and p-PTEN markers. The histopathological explanations were observed to be in line with biochemical analysis. Therefore, the finding of current work was that nimbolide has a defensive effect on the myocardium against DOX-induced cardiac tissue damage.
Collapse
Affiliation(s)
- Haining Li
- Department of Critical Care Medicine, Shenyang Tenth People's Hospital, Shenyang, Liaoning, China
| | - Bihua Xia
- Internal Medicine-Cardiovascular Department, The Second Affiliated Hospital of GuiZhou Medical University, Kaili, GuiZhou, China
| | - Wei Chen
- Department of Critical Care Medicine, Shenyang Tenth People's Hospital, Shenyang, Liaoning, China
| | - Yumeng Zhang
- Department of Critical Care Medicine, Shenyang Tenth People's Hospital, Shenyang, Liaoning, China
| | - Xia Gao
- Ultrasonic Room, Shenyang Tenth People's Hospital, Shenyang, Liaoning, China
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Sulaiman A Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Yujie Zhao
- ICU, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
73
|
Maayah ZH, Takahara S, Dyck JRB. The beneficial effects of reducing NLRP3 inflammasome activation in the cardiotoxicity and the anti-cancer effects of doxorubicin. Arch Toxicol 2020; 95:1-9. [PMID: 32852568 DOI: 10.1007/s00204-020-02876-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 08/12/2020] [Indexed: 01/07/2023]
Abstract
Doxorubicin (DOX) is a powerful broad-spectrum anti-neoplastic anthracycline antibiotic. However, DOX may cause a dose-dependent cardiotoxicity that can eventually progress to congestive heart failure and death. Numerous molecular mechanisms have been implicated in the cardiotoxic effect of DOX including topoisomerase IIβ and generation of free radicals. However, targeting these pathways appears to be insufficient to mitigate the cardiotoxic effects of DOX and/or ultimately reduces the anti-tumor activity of DOX. Thus, there remains a crucial need to identify novel pharmacological targets that can alleviate the cardiotoxic effects of DOX without reducing its anti-tumor activity. Recent studies have suggested that the Nucleotide-Binding Domain-Like Receptor Protein 3 (NLRP3) inflammasome is implicated in tumor progression and the chemoresistance of cancer cells to DOX. Of interest, reducing NLRP3 inflammasome activity alleviates DOX-induced cardiotoxicity. Therefore, we postulate that strategies that target the NLRP3 inflammasome can help mitigate the cardiotoxic effects of DOX while maintaining and/or even enhancing its anti-cancer activity. Herein, we review the current knowledge about the potential implication of the NLRP3 inflammasome in the anti-cancer and cardiotoxic effects of DOX.
Collapse
Affiliation(s)
- Zaid H Maayah
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Shingo Takahara
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Jason R B Dyck
- Cardiovascular Research Centre, Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada. .,458 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| |
Collapse
|
74
|
Ma P, Qin Y, Cao H, Erben U, Ni C, Qin Z. Temporary blockade of interferon-γ ameliorates doxorubicin-induced cardiotoxicity without influencing the anti-tumor effect. Biomed Pharmacother 2020; 130:110587. [PMID: 32763819 DOI: 10.1016/j.biopha.2020.110587] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/26/2020] [Accepted: 07/28/2020] [Indexed: 11/25/2022] Open
Abstract
Doxorubicin (DOX) is commonly used as an anti-cancer agent. However, its severe cardiotoxicity often makes it life threatening even long after DOX therapy during childhood. We recently reported interferon-γ (IFN-γ) necessary for DOX-induced acute cardiotoxicity in a p38 dependent way and, asked here for the potential of IFN-γ blockade to prevent DOX-induced chronic cardiotoxicity during tumor therapy. In our model system, mice without or with growing tumors repeatedly received DOX treatment. Simultaneous injection of anti-IFN-γ antibody R46-A2 with DOX to block IFN-γ signal efficiently protected the cardiac function of DOX treated recipients. Importantly, a single late injection of R46-A2 after DOX exposure also ameliorated DOX induced cardiac dysfunction in tumor-bearing mice. The anti-IFN-γ treatment did not affect the DOX-mediated tumor suppression effect and it left the main cellular immune response intact. Therefore, temporary blockade of IFN-γ may represent a novel strategy to ameliorate established DOX induced cardiotoxicity (DIC) or prevent its development in tumor therapy.
Collapse
Affiliation(s)
- Pan Ma
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Yue Qin
- National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | - Hong Cao
- Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ulrike Erben
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chen Ni
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China.
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, 450052, China; Key Laboratory of Protein and Peptide Pharmaceuticals, CAS-University of Tokyo Joint Laboratory of Structural Virology and Immunology, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
75
|
Tocchetti CG, Ameri P, de Boer RA, D’Alessandra Y, Russo M, Sorriento D, Ciccarelli M, Kiss B, Bertrand L, Dawson D, Falcao-Pires I, Giacca M, Hamdani N, Linke WA, Mayr M, van der Velden J, Zacchigna S, Ghigo A, Hirsch E, Lyon AR, Görbe A, Ferdinandy P, Madonna R, Heymans S, Thum T. Cardiac dysfunction in cancer patients: beyond direct cardiomyocyte damage of anticancer drugs: novel cardio-oncology insights from the joint 2019 meeting of the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart. Cardiovasc Res 2020; 116:1820-1834. [DOI: 10.1093/cvr/cvaa222] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/17/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
In western countries, cardiovascular (CV) disease and cancer are the leading causes of death in the ageing population. Recent epidemiological data suggest that cancer is more frequent in patients with prevalent or incident CV disease, in particular, heart failure (HF). Indeed, there is a tight link in terms of shared risk factors and mechanisms between HF and cancer. HF induced by anticancer therapies has been extensively studied, primarily focusing on the toxic effects that anti-tumour treatments exert on cardiomyocytes. In this Cardio-Oncology update, members of the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart discuss novel evidence interconnecting cardiac dysfunction and cancer via pathways in which cardiomyocytes may be involved but are not central. In particular, the multiple roles of cardiac stromal cells (endothelial cells and fibroblasts) and inflammatory cells are highlighted. Also, the gut microbiota is depicted as a new player at the crossroads between HF and cancer. Finally, the role of non-coding RNAs in Cardio-Oncology is also addressed. All these insights are expected to fuel additional research efforts in the field of Cardio-Oncology.
Collapse
Affiliation(s)
- Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, AB31, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Yuri D’Alessandra
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Michele Russo
- Department of Translational Medical Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Michele Ciccarelli
- Department of Medicine Surgery and Odontology, University of Salerno, Salerno, Italy
| | - Bernadett Kiss
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
| | - Luc Bertrand
- IREC Institute, Pole of Cardiovascular Research, Université Catholique de Louvain, Brussels, Belgium
| | - Dana Dawson
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Ines Falcao-Pires
- Unidade de Investigação e Desenvolvimento Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Portugal
| | - Mauro Giacca
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Ruhr Universität Bochum, Bochum, Germany
- Department of Cardiology, St. Joseph Hospital, Ruhr University Bochum, Witten, Germany
| | | | - Manuel Mayr
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences Institute, Amsterdam, The Netherlands
| | - Serena Zacchigna
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Imperial College London, London, UK
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Pisa, Italy
- Center for Cardiovascular Biology and Atherosclerosis Research, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| |
Collapse
|
76
|
The selective c-Met inhibitor capmatinib offsets cisplatin-nephrotoxicity and doxorubicin-cardiotoxicity and improves their anticancer efficacies. Toxicol Appl Pharmacol 2020; 398:115018. [DOI: 10.1016/j.taap.2020.115018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 02/06/2023]
|
77
|
Rocca C, Pasqua T, Cerra MC, Angelone T. Cardiac Damage in Anthracyclines Therapy: Focus on Oxidative Stress and Inflammation. Antioxid Redox Signal 2020; 32:1081-1097. [PMID: 31928066 DOI: 10.1089/ars.2020.8016] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Despite their serious side effects, anthracyclines (ANTs) are the most prescribed chemotherapeutic drugs because of their strong efficacy in both solid and hematological tumors. A major limitation to ANTs clinical application is the severe cardiotoxicity observed both acutely and chronically. The mechanism underlying cardiac dysfunction under chemotherapy is mainly dependent on the generation of oxidative stress and systemic inflammation, both of which lead to progressive cardiomyopathy and heart failure. Recent Advances: Over the years, the iatrogenic ANTs-induced cardiotoxicity was believed to be simply given by iron metabolism and reactive oxygen species production; however, several experimental data indicate that ANTs may use alternative damaging mechanisms, such as topoisomerase 2β inhibition, inflammation, pyroptosis, immunometabolism, and autophagy. Critical Issues: In this review, we aimed at discussing ANTs-induced cardiac injury from different points of view, updating and focusing on oxidative stress and inflammation, since these pathways are not exclusive or independent from each other but they together importantly contribute to the complexity of ANTs-induced multifactorial cardiotoxicity. Future Directions: A deeper understanding of the mechanistic signaling leading to ANTs side effects could reveal crucial targeting molecules, thus representing strategic knowledge to promote better therapeutic efficacy and lower cardiotoxicity during clinical application.
Collapse
Affiliation(s)
- Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Teresa Pasqua
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy
| | - Maria Carmela Cerra
- Laboratory of Organ and System Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences, University of Calabria, Rende, Italy.,National Institute of Cardiovascular Research (INRC), Bologna, Italy
| |
Collapse
|
78
|
Zeng C, Duan F, Hu J, Luo B, Huang B, Lou X, Sun X, Li H, Zhang X, Yin S, Tan H. NLRP3 inflammasome-mediated pyroptosis contributes to the pathogenesis of non-ischemic dilated cardiomyopathy. Redox Biol 2020; 34:101523. [PMID: 32273259 PMCID: PMC7327979 DOI: 10.1016/j.redox.2020.101523] [Citation(s) in RCA: 243] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/18/2020] [Accepted: 03/24/2020] [Indexed: 12/20/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is one of the most common causes of heart failure, and the underlying mechanism remains largely elusive. Here we investigated whether NLRP3 inflammasome-mediated pyroptosis contributes to non-ischemic DCM and dissected the underlying mechanism. We found that hyper activated NLRP3 inflammasome with pyroptotic cell death of cardiomyocytes were presented in the myocardial tissues of DCM patients, which were negatively correlated with cardiac function. Doxorubicin (Dox)-induced DCM characterization disclosed that NLRP3 inflammasome activation and pyroptosis occurred in Dox-treated heart tissues, but were very marginal in either NLRP3-/- or caspase-1-/- mice. Mechanistically, Dox enhanced expressions of NOX1 and NOX4 and induced mitochondrial fission through dynamin-related protein 1 (Drp1) activation, leading to NLRP3 inflammasome-mediated pyroptosis in cardiomyocytes via caspase-1-dependent manner. Conversely, both inhibitions of NOX1 and NOX4 and Drp1 suppressed Dox-induced NLPR3 inflammasome activation and pyroptosis. The alterations of NOX1 and NOX4 expression, Drp1 phosphorylation and mitochondrial fission were validated in DCM patients and mice. Importantly, Dox-induced Drp1-mediated mitochondrial fission and the consequent NLRP3 inflammasome activation and pyroptosis were reversed by NOX1 and NOX4 inhibition in mice. This study demonstrates for the first time that cardiomyocyte pyroptosis triggered by NLRP3 inflammasome activation via caspase-1 causally contributes to myocardial dysfunction progression and DCM pathogenesis.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Fengqi Duan
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jia Hu
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Bin Luo
- Department of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Binlong Huang
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaoying Lou
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510655, China
| | - Xiuting Sun
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Hongyu Li
- Laboratory Animal Center, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xuanhong Zhang
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shengli Yin
- Department of Cardiac Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| | - Hongmei Tan
- Department of Pathophysiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China; Guangdong Engineering & Technology Research Center for Disease-Model Animals, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
79
|
Morita Y, Leslie M, Kameyama H, Lokesh GLR, Ichimura N, Davis R, Hills N, Hasan N, Zhang R, Kondo Y, Gorenstein DG, Volk DE, Chervoneva I, Rui H, Tanaka T. Functional Blockade of E-Selectin in Tumor-Associated Vessels Enhances Anti-Tumor Effect of Doxorubicin in Breast Cancer. Cancers (Basel) 2020; 12:cancers12030725. [PMID: 32204492 PMCID: PMC7140021 DOI: 10.3390/cancers12030725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/11/2020] [Accepted: 03/11/2020] [Indexed: 12/26/2022] Open
Abstract
Chemotherapy is a mainstay of treatment for solid tumors. However, little is known about how therapy-induced immune cell infiltration may affect therapy response. We found substantial CD45+ immune cell density adjacent to E-selectin expressing inflamed vessels in doxorubicin (DOX)-treated residual human breast tumors. While CD45 level was significantly elevated in DOX-treated wildtype mice, it remained unchanged in DOX-treated tumors from E-selectin null mice. Similarly, intravenous administration of anti-E-selectin aptamer (ESTA) resulted in a significant reduction in CD45+ immune cell density in DOX-treated residual tumors, which coincided with a delay in tumor growth and lung metastasis in MMTV-pyMT mice. Additionally, both tumor infiltrating T-lymphocytes and tumor associated-macrophages were skewed towards TH2 in DOX-treated residual breast tumors; however, ESTA suppressed these changes. This study suggests that DOX treatment instigates de novo intratumoral infiltration of immune cells through E-selectin, and functional blockade of E-selectin may reduce residual tumor burden as well as metastasis through suppression of TH2 shift.
Collapse
Affiliation(s)
- Yoshihiro Morita
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE, 10th, Oklahoma City, OK 73104, USA; (Y.M.); (M.L.); (H.K.); (N.I.)
| | - Macall Leslie
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE, 10th, Oklahoma City, OK 73104, USA; (Y.M.); (M.L.); (H.K.); (N.I.)
| | - Hiroyasu Kameyama
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE, 10th, Oklahoma City, OK 73104, USA; (Y.M.); (M.L.); (H.K.); (N.I.)
| | - Ganesh L. R. Lokesh
- McGovern Medical School, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Hermann Pressler, Houston, TX 77030, USA; (G.L.R.L.); (D.E.V.)
| | - Norihisa Ichimura
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE, 10th, Oklahoma City, OK 73104, USA; (Y.M.); (M.L.); (H.K.); (N.I.)
| | - Rachel Davis
- School of Medicine, University of Oklahoma Health Sciences Center, 800 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA; (R.D.); (N.H.)
| | - Natalie Hills
- School of Medicine, University of Oklahoma Health Sciences Center, 800 Stanton L. Young Blvd., Oklahoma City, OK 73104, USA; (R.D.); (N.H.)
| | - Nafis Hasan
- Department of Pharmaceutical Sciences, Thomas Jefferson University, 1020 Locust St, Philadelphia, PA 19107, USA;
| | - Roy Zhang
- Department of Pathology, College of Medicine, University of Oklahoma Health Sciences Center, 940 SL Young Blvd, Oklahoma City, OK 73104, USA;
| | - Yuji Kondo
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, 825 NE. 13th, Oklahoma City, OK 73104, USA;
| | | | - David E. Volk
- McGovern Medical School, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, 1825 Hermann Pressler, Houston, TX 77030, USA; (G.L.R.L.); (D.E.V.)
| | - Inna Chervoneva
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, 1015 Chestnut St., Philadelphia, PA 19107, USA;
| | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI 53226, USA;
| | - Takemi Tanaka
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, 975 NE, 10th, Oklahoma City, OK 73104, USA; (Y.M.); (M.L.); (H.K.); (N.I.)
- Department of Pathology, College of Medicine, University of Oklahoma Health Sciences Center, 940 SL Young Blvd, Oklahoma City, OK 73104, USA;
- Correspondence: ; Tel.: +1-(405)-271-8260
| |
Collapse
|
80
|
Phungphong S, Kijtawornrat A, Kampaengsri T, Wattanapermpool J, Bupha-Intr T. Comparison of exercise training and estrogen supplementation on mast cell-mediated doxorubicin-induced cardiotoxicity. Am J Physiol Regul Integr Comp Physiol 2020; 318:R829-R842. [PMID: 32159365 DOI: 10.1152/ajpregu.00224.2019] [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] [Indexed: 12/14/2022]
Abstract
Cardiac inflammation has been proposed as one of the primary mechanisms of anthracycline-induced acute cardiotoxicity. A reduction in cardiac inflammation might also reduce cardiotoxicity. This study aimed to evaluate the potential of estrogen therapy and regular exercise on attenuating cardiac inflammation in the context of doxorubicin-induced cardiomyopathy. Ovariectomized rats were randomly allocated into estrogen supplementation, exercise training, and mast cell stabilizer treatment groups. Eight weeks after ovariectomy, rats received six cumulative doses of doxorubicin for two weeks. Echocardiography demonstrated a progressive decrease in ejection fraction in doxorubicin-treated rats without hypertrophic effect. This systolic defect was completely prevented by either estrogen supplementation or mast cell stabilizer treatment but not by regular exercise. As a heart disease indicator, increased β-myosin heavy chain expression induced by doxorubicin could only be prevented by estrogen supplementation. Decrease in shortening and intracellular Ca2+ transients of cardiomyocytes were due to absence of female sex hormones without further effects of doxorubicin. Again, estrogen supplementation and mast cell stabilizer treatment prevented these changes but exercise training did not. Histological analysis indicated that the hyperactivation of cardiac mast cells in ovariectomized rats was augmented by doxorubicin. Estrogen supplementation and mast cell stabilizer treatment completely prevented both increases in mast cell density and degranulation, whereas exercise training partially attenuated the hyperactivation. Our results, therefore, suggest that estrogen supplementation acts similarly to mast cell stabilizers in attenuating the effects of doxorubicin. Ineffectiveness of regular exercise in preventing the acute cardiotoxicity of doxorubicin might be due to a lesser effect on preventing cardiac inflammation.
Collapse
Affiliation(s)
- Sukanya Phungphong
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Anusak Kijtawornrat
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | | | | | - Tepmanas Bupha-Intr
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
81
|
Sun Z, Lu W, Lin N, Lin H, Zhang J, Ni T, Meng L, Zhang C, Guo H. Dihydromyricetin alleviates doxorubicin-induced cardiotoxicity by inhibiting NLRP3 inflammasome through activation of SIRT1. Biochem Pharmacol 2020; 175:113888. [PMID: 32112883 DOI: 10.1016/j.bcp.2020.113888] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 02/24/2020] [Indexed: 12/21/2022]
Abstract
Doxorubicin (DOX) is a powerful anthracycline antineoplastic drug whose clinical application is limited by serious cardiotoxic side effects. Dihydromyricetin (DHM), a flavonoid compound extracted from the Japanese raisin tree (Hovenia dulcis), is cardioprotective in patients with heart failure; however, the underlying mechanisms are poorly understood. The aim of this study was to assess the possible anti-inflammatory properties of DHM in a rat model of DOX-induced cardiotoxicity and DOX-treated H9C2 cells, and gain insights into the molecular mechanisms that mediate these effects. The results showed that DHM treatment significantly improved the myocardial structure and function in DOX-exposed rats by alleviating NLRP3 inflammasome-mediated inflammation. DHM also inhibited DOX-induced activation of the NLRP3 inflammasome in H9C2 cells. This effect was mediated by inhibition of caspase-1 activity, suppression of IL-1β and IL-18 release, and upregulation of SIRT1 protein levels in vivo and in vitro. Moreover, selective inhibition of SIRT1 blocked the protective effects of DHM. Collectively, our findings indicate that DHM protects against DOX-induced cardiotoxicity by inhibiting NLRP3 inflammasome activation via stimulation of the SIRT1 pathway.
Collapse
Affiliation(s)
- Zhenzhu Sun
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China
| | - Wenqiang Lu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Na Lin
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Hui Lin
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | - Jie Zhang
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | - Tingjuan Ni
- Zhejiang University School of Medicine, Hangzhou, China
| | - Liping Meng
- Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China
| | | | - Hangyuan Guo
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou 325000, Zhejiang, China; Department of Cardiology, Shaoxing People's Hospital, Shaoxing Hospital of Zhejiang University, Shaoxing 312000, Zhejiang, China.
| |
Collapse
|
82
|
Grant MK, Abdelgawad IY, Lewis CA, Zordoky BN. Sexual Dimorphism in Doxorubicin-induced Systemic Inflammation: Implications for Hepatic Cytochrome P450 Regulation. Int J Mol Sci 2020; 21:ijms21041279. [PMID: 32074957 PMCID: PMC7072970 DOI: 10.3390/ijms21041279] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/03/2020] [Accepted: 02/11/2020] [Indexed: 12/20/2022] Open
Abstract
Doxorubicin (DOX) is an effective chemotherapeutic agent used to treat a wide variety of malignancies. In addition to its multi-organ toxicity, DOX treatment has been shown to induce systemic inflammation in patients and experimental animals. Inflammation alters the expression of hepatic cytochrome P450 (CYP) enzymes, which play important roles in drug metabolism and DOX-induced toxicity. Significant sex differences have been reported in DOX-induced toxicity; however, sex differences in DOX-induced systemic inflammation and the potential effects on hepatic CYP expression have not been determined. In the current work, male and female C57Bl/6 mice were administered DOX (20 mg/kg by intraperitoneal injection), and groups of mice were sacrificed 24 and 72 h after DOX administration. DOX elicited a systemic inflammatory response in both male and female mice, but the inflammatory response was stronger in male mice. DOX altered the expression of hepatic CYP isoforms in a sex-dependent manner. Most notably, inhibition of Cyp2c29 and Cyp2e1 was stronger in male than in female mice, which paralleled the sex differences in systemic inflammation. Therefore, sex differences in DOX-induced systemic inflammation may lead to sexually dimorphic drug interactions, in addition to contributing to the previously reported sexual dimorphism in specific DOX-induced organ toxicity.
Collapse
|
83
|
Teixeira AAS, Biondo LA, Silveira LS, Lima EA, Batatinha HA, Diniz TA, Oliveira De Souza C, Comin J, Neto JCR. Doxorubicin modulated clock genes and cytokines in macrophages extracted from tumor-bearing mice. Cancer Biol Ther 2020; 21:344-353. [PMID: 31931676 PMCID: PMC7515505 DOI: 10.1080/15384047.2019.1702400] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Circadian rhythm is essential for cellular regulation of physiological, metabolic, and immune functions. Perturbations of circadian rhythms have been correlated with increased susceptibility to cancer and poor prognosis in the cancer treatment. Our aim is to investigate the role of doxorubicin (DOX) treatment on clock genes expression and inflammation in intraperitoneal macrophages and the antitumoral response. Methods: Macrophages were extracted from intraperitoneal cavity of mice without or with Lewis lung carcinoma (LLC) and treated with DOX totaling four groups (CTL, LLC, LLC+DOX and DOX) and analyzes of clock genes in six time points (ZT02, ZT06, ZT10, ZT14, ZT18 AND ZT22). Intraperitoneal macrophages cell culture was stimulated with LPS and DOX and clock genes and inflammatory profile were analyzed. In tumor were analyzed macrophages markers. Results: The expression of F4/80 (ZT22) and CD11c (ZT06) tumor tissue was significantly differed between LLC and LCC+DOX groups. In the intraperitoneal macrophages, DOX increased Clock (ZT10), Rev-Erbα (ZT18 and ZT22) and Per2 expressions (ZT18); in the LLC+DOX group was increased Bmal1 (ZT10), Per2 (ZT18) and NF-kB (ZT22) expressions; IL-6 expression increased in the LCC group (ZT02). In intraperitoneal macrophages cell culture stimulated with DOX and LPS after 24 h decreased Clock and Per1. DOX causes depression after 6 and 24 h in TNF-α content and Per2 gene expression after 24 h IL-1β expression was reduced also. Conclusion: DOX treatment in vivo disrupted cytokine and clock genes expression in intraperitoneal macrophages suppressing immune response. Moreover, macrophages cultured with DOX had decreased expression of LPS-stimulated inflammatory cytokines.
Collapse
Affiliation(s)
| | | | - Loreana S Silveira
- Institute of BiomedicalSciences, University of São Paulo, São Paulo, Brazil
| | - Edson A Lima
- Institute of BiomedicalSciences, University of São Paulo, São Paulo, Brazil
| | - Helena A Batatinha
- Institute of BiomedicalSciences, University of São Paulo, São Paulo, Brazil
| | - Tiego A Diniz
- Institute of BiomedicalSciences, University of São Paulo, São Paulo, Brazil
| | | | - Jeferson Comin
- Institute of BiomedicalSciences, University of São Paulo, São Paulo, Brazil
| | | |
Collapse
|
84
|
Vince JE, De Nardo D, Gao W, Vince AJ, Hall C, McArthur K, Simpson D, Vijayaraj S, Lindqvist LM, Bouillet P, Rizzacasa MA, Man SM, Silke J, Masters SL, Lessene G, Huang DCS, Gray DHD, Kile BT, Shao F, Lawlor KE. The Mitochondrial Apoptotic Effectors BAX/BAK Activate Caspase-3 and -7 to Trigger NLRP3 Inflammasome and Caspase-8 Driven IL-1β Activation. Cell Rep 2019; 25:2339-2353.e4. [PMID: 30485804 DOI: 10.1016/j.celrep.2018.10.103] [Citation(s) in RCA: 160] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 08/08/2018] [Accepted: 10/26/2018] [Indexed: 11/25/2022] Open
Abstract
Intrinsic apoptosis resulting from BAX/BAK-mediated mitochondrial membrane damage is regarded as immunologically silent. We show here that in macrophages, BAX/BAK activation results in inhibitor of apoptosis (IAP) protein degradation to promote caspase-8-mediated activation of IL-1β. Furthermore, BAX/BAK signaling induces a parallel pathway to NLRP3 inflammasome-mediated caspase-1-dependent IL-1β maturation that requires potassium efflux. Remarkably, following BAX/BAK activation, the apoptotic executioner caspases, caspase-3 and -7, act upstream of both caspase-8 and NLRP3-induced IL-1β maturation and secretion. Conversely, the pyroptotic cell death effectors gasdermin D and gasdermin E are not essential for BAX/BAK-induced IL-1β release. These findings highlight that innate immune cells undergoing BAX/BAK-mediated apoptosis have the capacity to generate pro-inflammatory signals and provide an explanation as to why IL-1β activation is often associated with cellular stress, such as during chemotherapy.
Collapse
Affiliation(s)
- James E Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| | - Dominic De Nardo
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Wenqing Gao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Angelina J Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Cathrine Hall
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
| | - Kate McArthur
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Daniel Simpson
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Swarna Vijayaraj
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Lisa M Lindqvist
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Philippe Bouillet
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Mark A Rizzacasa
- School of Chemistry, The Bio 21 Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Si Ming Man
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, Australian National University, Canberra 2601, Australia
| | - John Silke
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Seth L Masters
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Guillaume Lessene
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - David C S Huang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Daniel H D Gray
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Benjamin T Kile
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Feng Shao
- National Institute of Biological Sciences, Beijing 102206, China
| | - Kate E Lawlor
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
85
|
Liu P, Lu Z, Liu L, Li R, Liang Z, Shen M, Xu H, Ren D, Ji M, Yuan S, Shang D, Zhang Y, Liu H, Tu Z. NOD-like receptor signaling in inflammation-associated cancers: From functions to targeted therapies. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 64:152925. [PMID: 31465982 DOI: 10.1016/j.phymed.2019.152925] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/06/2019] [Accepted: 04/08/2019] [Indexed: 05/22/2023]
Abstract
BACKGROUND Recently, many studies have reported that some botanicals and natural products were able to regulate NOD-like receptor signaling. NOD-like receptors (NLRs) have been established as crucial regulators in inflammation-associated tumorigenesis, angiogenesis, cancer cell stemness and chemoresistance. NLRs specifically sense pathogen-associated molecular patterns and respond by activating other signaling regulators, including Rip2 kinase, NF-κB, MAPK and ASC/caspase-1, leading to the secretion of various cytokines. PURPOSE The aim of this article is to review the molecular mechanisms of NOD-like receptor signaling in inflammation-associated cancers and the NLRs-targeted botanicals and synthetic small molecules in cancer intervention. RESULTS Aberrant activation of NLRs occurs in various cancers, orchestrating the tissue microenvironment and potentiating neoplastic risk. Blocking NLR inflammasome activation by botanicals or synthetic small molecules may be a valuable way to prevent cancer progression. Moreover, due to the roles of NLRs in regulating cytokine production, NLR signaling may be correlated with senescence-associated secretory phenotype. CONCLUSION In this review, we discuss how NLR signaling is involved in inflammation-associated cancers, and highlight the NLR-targeted botanicals and synthetic small molecules in cancer intervention.
Collapse
Affiliation(s)
- Peng Liu
- Institute of Life Sciences, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China
| | - Ziwen Lu
- School of Pharmacy, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China
| | - Lanlan Liu
- Institute of Life Sciences, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China
| | - Ruyan Li
- Institute of Life Sciences, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China
| | - Zhiquan Liang
- School of Pharmacy, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China
| | - Mingxiang Shen
- Institute of Life Sciences, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China
| | - Han Xu
- School of Pharmacy, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China
| | - Dewan Ren
- School of Pharmacy, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China
| | - Mengchen Ji
- Institute of Life Sciences, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China
| | - Sirui Yuan
- School of Pharmacy, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China
| | - Dongsheng Shang
- School of Pharmacy, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China
| | - Yibang Zhang
- School of Pharmacy, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China
| | - Hanqing Liu
- School of Pharmacy, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China.
| | - Zhigang Tu
- Institute of Life Sciences, Jiangsu University, 301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
86
|
Pal S, Medatwal N, Kumar S, Kar A, Komalla V, Yavvari PS, Mishra D, Rizvi ZA, Nandan S, Malakar D, Pillai M, Awasthi A, Das P, Sharma RD, Srivastava A, Sengupta S, Dasgupta U, Bajaj A. A Localized Chimeric Hydrogel Therapy Combats Tumor Progression through Alteration of Sphingolipid Metabolism. ACS CENTRAL SCIENCE 2019; 5:1648-1662. [PMID: 31660434 PMCID: PMC6813554 DOI: 10.1021/acscentsci.9b00551] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Indexed: 05/14/2023]
Abstract
Rapid proliferation of cancer cells assisted by endothelial cell-mediated angiogenesis and acquired inflammation at the tumor microenvironment (TME) lowers the success rate of chemotherapeutic regimens. Therefore, targeting these processes using localized delivery of a minimally toxic drug combination may be a promising strategy. Here, we present engineering of a biocompatible self-assembled lithocholic acid-dipeptide derived hydrogel (TRI-Gel) that can maintain sustained delivery of antiproliferating doxorubicin, antiangiogenic combretastatin-A4 and anti-inflammatory dexamethasone. Application of TRI-Gel therapy to a murine tumor model promotes enhanced apoptosis with a concurrent reduction in angiogenesis and inflammation, leading to effective abrogation of tumor proliferation and increased median survival with reduced drug resistance. In-depth RNA-sequencing analysis showed that TRI-Gel therapy induced transcriptome-wide alternative splicing of many genes responsible for oncogenic transformation including sphingolipid genes. We demonstrate that TRI-Gel therapy targets the reversal of a unique intron retention event in β-glucocerebrosidase 1 (Gba1), thereby increasing the availability of functional Gba1 protein. An enhanced Gba1 activity elevates ceramide levels responsible for apoptosis and decreases glucosylceramides to overcome drug resistance. Therefore, TRI-Gel therapy provides a unique system that affects the TME via post-transcriptional modulations of sphingolipid metabolic genes, thereby opening a new and rational approach to cancer therapy.
Collapse
Affiliation(s)
- Sanjay Pal
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana, India
- Kalinga
Institute of Industrial Technology, Bhubaneswar 751024, Odisha, India
| | - Nihal Medatwal
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana, India
- Manipal
Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Sandeep Kumar
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana, India
- Manipal
Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Animesh Kar
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana, India
| | - Varsha Komalla
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana, India
| | - Prabhu Srinivas Yavvari
- Department
of Chemistry, Indian Institute of Science
Education and Research, Bhopal 462066, Madhya Pradesh, India
| | - Deepakkumar Mishra
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana, India
| | - Zaigham Abbas Rizvi
- Translational
Health Science and Technology
Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana, India
| | - Shiv Nandan
- Amity Institute
of Integrative Sciences and Health, Amity
University Haryana, Panchgaon, Manesar, Gurgaon 122413, Haryana, India
| | - Dipankar Malakar
- SCIEX, 121 Udyog Vihar,
Phase IV, Gurgaon 122015, Haryana, India
| | - Manoj Pillai
- SCIEX, 121 Udyog Vihar,
Phase IV, Gurgaon 122015, Haryana, India
| | - Amit Awasthi
- Translational
Health Science and Technology
Institute, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana, India
| | - Prasenjit Das
- Department
of Pathology, All India Institute of Medical
Sciences, Ansari Nagar, New Delhi 110029, India
| | - Ravi Datta Sharma
- Amity Institute
of Integrative Sciences and Health, Amity
University Haryana, Panchgaon, Manesar, Gurgaon 122413, Haryana, India
| | - Aasheesh Srivastava
- Department
of Chemistry, Indian Institute of Science
Education and Research, Bhopal 462066, Madhya Pradesh, India
| | - Sagar Sengupta
- National
Institute of Immunology, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Ujjaini Dasgupta
- Amity Institute
of Integrative Sciences and Health, Amity
University Haryana, Panchgaon, Manesar, Gurgaon 122413, Haryana, India
- E-mail: . (U.D.)
| | - Avinash Bajaj
- Laboratory
of Nanotechnology and Chemical Biology, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon
Expressway, Faridabad 121001, Haryana, India
- E-mail: . (A.B.)
| |
Collapse
|
87
|
Razazan A, Behravan J. Single peptides and combination modalities for triple negative breast cancer. J Cell Physiol 2019; 235:4089-4108. [PMID: 31642059 DOI: 10.1002/jcp.29300] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/27/2019] [Indexed: 12/31/2022]
Abstract
Unlike other types of breast cancers (BCs), no specific therapeutic targets have been established for triple negative breast cancer (TNBC). Therefore, chemotherapy and radiotherapy are the only available adjuvant therapeutic choices for TNBC. New emerging reports show that TNBC is associated with higher numbers of intratumoral tumor infiltrating lymphocytes. This is indicative of host anti-TNBC immune surveillance and suggesting that immunotherapy can be considered as a therapeutic approach for TNBC management. Recent progress in molecular mechanisms of tumor-immune system interaction and cancer vaccine development studies, fast discoveries and FDA approvals of immune checkpoint inhibitors, chimeric antigen receptor T-cells, and oncolytic virotherapy have significantly attracted attention and research directions toward the immunotherapeutic approach to TNBC. Here in this review different aspects of TNBC immunotherapies including the host immune system-tumor interactions, the tumor microenvironment, the relevant molecular targets for immunotherapy, and clinical trials in the field are discussed.
Collapse
Affiliation(s)
- Atefeh Razazan
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Center for Obesity, Diabetes and Metabolism (Internal Medicine-Molecular Medicine), Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Javad Behravan
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, University of Waterloo, Waterloo, Canada.,Theraphage Inc., Kitchener, Ontario, Canada
| |
Collapse
|
88
|
Muhammad JS, Jayakumar MN, Elemam NM, Venkatachalam T, Raju TK, Hamoudi RA, Maghazachi AA. Gasdermin D Hypermethylation Inhibits Pyroptosis And LPS-Induced IL-1β Release From NK92 Cells. Immunotargets Ther 2019; 8:29-41. [PMID: 31687364 PMCID: PMC6800286 DOI: 10.2147/itt.s219867] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 09/28/2019] [Indexed: 01/09/2023] Open
Abstract
Introduction Although natural killer (NK) are major cells used to treat cancer patients, recent clinical trials showed that NK92 cells can be also used for the same purpose due to their high anti-tumor activity. Here, we examined whether these cells might be inflammatory due to the release of interleukin-1β (IL-1β), and whether the anti-inflammatory molecules dimethyl fumarate (DMF), or monomethyl fumarate (MMF) impair this activity. Methods NK92 cells were examined for the synthesis and release of IL-1β utilizing RT-PCR and ELISA assay, respectively. The expression of hydroxy-carboxylic acid receptors (HCA)1, HCA2 and HCA3 was detected by immunoblotting, flow cytometry, immunofluorescence and RT-PCR assays. The activation of caspase-1 and Gasdermin D (GSDMD) was evaluated by immunoblot assay. Pyroptosis was demonstrated by immunofluorescence imaging. Expression of DNA methyltransferases (DNMTs) mRNA was determined by whole transcriptome and immunoblot analyses. Results LPS-induced the release of IL-1β from NK92 cells, whereas DMF or MMF inhibited this induction. The effect of these drugs was due to inhibiting the conversion of procaspase-1 into active caspase-1. NK92 cells highly expressed GSDMD, a pyroptotic-mediated molecule. However, LPS induced the distribution of GSDMD into the cell membranes, corroborated with the presence of pyroptotic bodies, an activity that was inhibited by DMF or MMF. These molecule also inhibited the generation of GSDMD through DNMT-mediated hypermethylation of the promoter region of GSDMD gene. These results were supported by increased expression of DNMTs mRNA as determined by whole transcriptome analysis. Discussion Our results are the first to show that NK92 cells utilize GSDMD pathway to release IL-1β. Further, DMF and MMF which were previously shown to enhance NK cell cytotoxicity, also inhibit the inflammatory effects of these cells, making them most suitable for treating cancer patients. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/ZT7DsG-nq0o
Collapse
Affiliation(s)
- Jibran Sualeh Muhammad
- College of Medicine, and the Immuno-Oncology Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Manju Nidagodu Jayakumar
- College of Medicine, and the Immuno-Oncology Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Noha Mousaad Elemam
- College of Medicine, and the Immuno-Oncology Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Thenmozhi Venkatachalam
- College of Medicine, and the Immuno-Oncology Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Tom Kalathil Raju
- College of Medicine, and the Immuno-Oncology Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Rifat Akram Hamoudi
- College of Medicine, and the Immuno-Oncology Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Azzam A Maghazachi
- College of Medicine, and the Immuno-Oncology Group, Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates
| |
Collapse
|
89
|
Benner B, Scarberry L, Stiff A, Duggan MC, Good L, Lapurga G, Butchar JP, Tridandapani S, Carson WE. Evidence for interaction of the NLRP3 inflammasome and Bruton's tyrosine kinase in tumor-associated macrophages: implications for myeloid cell production of interleukin-1beta. Oncoimmunology 2019; 8:1659704. [PMID: 31646085 PMCID: PMC6791459 DOI: 10.1080/2162402x.2019.1659704] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 01/05/2023] Open
Abstract
An inflammatory microenvironment has been shown to play an important role in the growth and metastasis of tumors. The NLRP3 inflammasome is a multi-protein complex of the innate immune system that is responsible for the production of the potent inflammatory cytokine IL-1β. Tumor- associated macrophages (TAM) are an expanded population of immune cells found in the tumor microenvironment that can promote the initiation and metastasis of tumor cells. Their presence has been correlated with disease burden, highlighting the therapeutic potential of targeting this population. However, to date clinically relevant pharmacologic strategies to target TAM remain elusive. Here, we show that in vitro generated TAM harbor NLRP3 inflammasome components and produce IL-1β. Ibrutinib, an irreversible inhibitor of Bruton's tyrosine kinase (BTK), is in clinical use for the treatment of B- cell malignancies. We report that BTK is expressed by human in vitro generated TAM and murine macrophages and that it physically associates with the NLRP3 inflammasome. Furthermore, ibrutinib is able to inhibit BTK phosphorylation in TAM generated in vitro. Treatment of TAM with ibrutinib significantly impaired the ability of these cells to produce IL-1β. The present study provides evidence that BTK physically associates with the NLRP3 inflammasome and that inhibition of BTK with ibrutinib can impair the production of IL-1β by in vitro generated TAM. Thus, ibrutinib could potentially be of clinical use in abrogating inflammation-associated cancer progression and the immune-suppressive effects of myeloid cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Brooke Benner
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Luke Scarberry
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Andrew Stiff
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Megan C. Duggan
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Logan Good
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Gabriella Lapurga
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | | | | | - William E. Carson
- Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
- Division of Surgical Oncology, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
90
|
Immunological consequences of chemotherapy: Single drugs, combination therapies and nanoparticle-based treatments. J Control Release 2019; 305:130-154. [DOI: 10.1016/j.jconrel.2019.04.020] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/09/2019] [Accepted: 04/14/2019] [Indexed: 02/07/2023]
|
91
|
Li W, He W, Xia P, Sun W, Shi M, Zhou Y, Zhu W, Zhang L, Liu B, Zhu J, Zhu Y, Zhou E, Sun M, Gao K. Total Extracts of Abelmoschus manihot L. Attenuates Adriamycin-Induced Renal Tubule Injury via Suppression of ROS-ERK1/2-Mediated NLRP3 Inflammasome Activation. Front Pharmacol 2019; 10:567. [PMID: 31191310 PMCID: PMC6548014 DOI: 10.3389/fphar.2019.00567] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 05/06/2019] [Indexed: 12/11/2022] Open
Abstract
Abelmoschus manihot (L.) Medik. (Malvaceae) is a herb used in traditional Chinese medicine to treat some kidney diseases. To date, the detailed mechanisms by which A. manihot improves some kinds of renal disease are not fully understood. In this study, we established Adriamycin-induced NRK-52E cells, the normal rat kidney epithelial cell line, injury, and Sprague-Dawley rats with Adriamycin-induced nephropathy to evaluate the role and mechanisms of total extracts of A. manihot flower (TEA) both in vitro and in vivo. We found that TEA ameliorated Adriamycin-induced cellular morphological changes, cell viability, and apoptosis through the suppression of protein oxidation and ERK1/2 signaling. However, this anti-oxidative stress role of TEA was independent of ROS inhibition. Adriamycin activated ERK1/2 signaling followed by activation of NLRP3 inflammasomes. TEA suppressed NLRP3 inflammasomes via inhibition of ERK1/2 signal transduction; decreased proteinuria and attenuated renal tubule lesions; and inhibited the expression of NLRP3 in tubules in rats with Adriamycin nephropathy. Collectively, TEA protects renal tubular cells against Adriamycin-induced tubule injury via inhibition of ROS-ERK1/2-NLRP3 inflammasomes.
Collapse
Affiliation(s)
- Wei Li
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Weiming He
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Ping Xia
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Wei Sun
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Ming Shi
- Division of Gerontology, The Third Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yao Zhou
- Department of Pathophysiology, Xuzhou Medical University, Xuzhou, China
| | - Weiwei Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Lu Zhang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Buhui Liu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Jingjing Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Yiye Zhu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Enchao Zhou
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| | - Minjie Sun
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | - Kun Gao
- Affiliated Hospital of Nanjing University of Chinese Medicine, Division of Nephrology, Jiangsu Province Hospital of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
92
|
Son S, Shim DW, Hwang I, Park JH, Yu JW. Chemotherapeutic Agent Paclitaxel Mediates Priming of NLRP3 Inflammasome Activation. Front Immunol 2019; 10:1108. [PMID: 31156650 PMCID: PMC6532018 DOI: 10.3389/fimmu.2019.01108] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/01/2019] [Indexed: 12/19/2022] Open
Abstract
Paclitaxel is a chemotherapeutic drug commonly used to treat different types of cancer. In addition to its antitumor effect, paclitaxel is also known to promote Toll-like receptor (TLR) 4-dependent inflammatory responses, which may lower its chemotherapeutic efficacy. However, it remains unclear whether paclitaxel is able to affect inflammasome signaling in myeloid or cancer cells. Therefore, we examined the potential effect of paclitaxel on the activation of an inflammasome complex by examining caspase-1 activation and interleukin (IL)-1β secretion in bone marrow-derived macrophages (BMDMs). The results showed that treatment with paclitaxel alone or following LPS priming failed to trigger the secretion of active caspase-1 and IL-1β from BMDMs. However, paclitaxel could induce robust activation of caspase-1 in BMDMs in the presence of NLRP3 inflammasome-activating signal 2, such as ATP or nigericin. This paclitaxel/ATP-mediated inflammasome activation was completely abrogated in Nlrp3-deficient macrophages. Mechanistically, paclitaxel treatment induced robust activation of the TLR4 signaling cascade, including phosphorylation of IκB and JNK and upregulation of proinflammatory cytokine mRNA levels in a TLR4-dependent manner. In contrast, paclitaxel treatment alone did not induce mitochondrial damages such as the loss of the mitochondrial membrane potential and production of mitochondrial ROS. These findings suggest that paclitaxel can drive the priming of signal-mediated events for NLRP3 activation but not a second signal-triggered phenomenon such as mitochondrial damage. This suggestion was supported by the observations that paclitaxel treatment caused robust IL-1β production in macrophages in the presence of cell-free medium derived from growth of injured cells and also in the spleen of mice. Collectively, our data strongly indicate that paclitaxel is able to facilitate the activation of NLRP3 inflammasome signaling in a certain physiological environment.
Collapse
Affiliation(s)
- Seunghwan Son
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Do-Wan Shim
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Inhwa Hwang
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Jong-Hwan Park
- BK 21 PLUS Project Team, Laboratory Animal Medicine, College of Veterinary Medicine, Chonnam National University, Gwangju, South Korea
| | - Je-Wook Yu
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
93
|
Shati AA, El-Kott AF. Acylated ghrelin prevents doxorubicin-induced cardiac intrinsic cell death and fibrosis in rats by restoring IL-6/JAK2/STAT3 signaling pathway and inhibition of STAT1. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:1151-1168. [PMID: 31093684 DOI: 10.1007/s00210-019-01664-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/02/2019] [Indexed: 01/12/2023]
Abstract
This study investigated if JAK/STAT signaling pathway mediates doxorubicin (DOX)-induced cell death and fibrosis in left ventricles (LVs) of rats and examined if acylated ghrelin affords protection by modulating this pathway. Male rats (120 ± 5 g) were divided into 6 groups (10 rats each) as follows: control; control + AG (10 ng/kg, s.c.); DOX (an accumulative dose 15 mg/kg, i.p.); DOX + AG, DOX + AG + AG490, a JAK2 inhibitor (5 mg/kg, i.p.); and DOX + AG + [D-Lys3]-GHRP-6; an AG receptor antagonist (3.75 mg/kg, i.p.). All treatments were carried out for 35 days. In rats' LVs, DOX significantly impaired the systolic and diastolic functions, enhanced levels of ROS and MDA, reduced levels of GSH and Bcl-2, and increased mRNA and protein levels of collagen I/III and TGF-β and cleaved caspase-3. In addition, although DOX did not affect JAK1 or JAK2 activity, it significantly increased protein levels of IL-6, decreased STAT3 and p-STAT3 (Tyr701&Ser727), and increased STAT1 and p-STAT1 (Tyr701&Ser727) levels, with a concomitant decrease in ERK1/2 activity and an increase in P38 activity. However, without affecting IL-6 and JAK1/2, AG reversed all of the observed alterations with a significant increase in the levels and activities of JAK2. Similar effects of AG were also seen in control rats. Interestingly, all the beneficial effects afforded by AG were abolished by AG490 and AG + [D-Lys3]-GHRP-6. In conclusion, DOX-induced cardiac toxicity involves stimulation of IL-6, P38, and STAT1 signaling levels whereas the protective effect afforded by AG involves the activation of ERK1/2 and JAK2/STAT3 and inhibition of STAT1.
Collapse
Affiliation(s)
- Ali A Shati
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia.
| | - Attalla Farag El-Kott
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia.,Zoology Department, College of Science, Damanhour University, Damanhour, Egypt
| |
Collapse
|
94
|
Caubet Fernandez M, Drouin S, Samoilenko M, Morel S, Krajinovic M, Laverdière C, Sinnett D, Levy E, Marcil V, Lefebvre G. A Bayesian multivariate latent t-regression model for assessing the association between corticosteroid and cranial radiation exposures and cardiometabolic complications in survivors of childhood acute lymphoblastic leukemia: a PETALE study. BMC Med Res Methodol 2019; 19:100. [PMID: 31088361 PMCID: PMC6515639 DOI: 10.1186/s12874-019-0725-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 04/05/2019] [Indexed: 01/19/2023] Open
Abstract
Background Childhood acute lymphoblastic leukemia (cALL) is the most frequent pediatric cancer. Over the past decades, treatment of cALL has significantly improved, with cure rates close to 90%. However intensive chemotherapy and cranial radiotherapy (CRT) during a critical period of a child’s development have been shown to lead to significant long-term side effects including cardiometabolic complications. Using the PETALE (Prévenir les effets tardifs des traitements de la leucémie aiguë lymphoblastique chez l’enfant) cALL survivor cohort, we investigated the association between combined cumulative corticosteroids (CS) doses and CRT exposures and obesity, insulin resistance, (pre-)hypertension, and dyslipidemia jointly. Methods A Bayesian multivariate latent-t model which accounted for our correlated binary outcomes was used for the analyses (n = 241 survivors). CS doses were categorized as low (LD) or high (HD). Combined exposure levels investigated were: 1) LD/no CRT; 2) LD/CRT, and; 3) HD/CRT. We also performed complementary sensitivity analyses for covariate adjustment. Results Prevalence of cardiometabolic complications ranged from 12.0% for (pre-)hypertension to 40.2% for dyslipidemia. The fully adjusted odds ratio (OR) for dyslipidemia associated with LD/CRT (vs. LD/No CRT) was OR = 1.98 (95% credible interval (CrI): 1.02 to 3.88). LD/CRT level also led to a 0.15 (95% CrI: 0.00 to 0.29) excess risk to develop at least one cardiometabolic complication. Except for obesity, adjusted results for the highest exposure category HD/CRT were generally similar to those for LD/CRT albeit not statistically significant. White blood cell count at diagnosis, a proxy for cALL burden at diagnosis, was found associated with insulin resistance (OR = 1.08 for a 10-unit increase (× 109/L), 95% CrI: 1.02 to 1.14). Conclusions Our results indicated that combined LD/CRT exposure is a likely determinant of dyslipidemia among cALL survivors. No evidence was found to suggest that high doses of CS lead to additional risk for obesity, insulin resistance, (pre-)hypertension, and dyslipidemia beyond that induced by CRT. The multivariate model selected for analyses was judged globally useful to assess potential exposure-related concomitance of binary outcomes. Electronic supplementary material The online version of this article (10.1186/s12874-019-0725-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Miguel Caubet Fernandez
- Department of Mathematics, University of Quebec at Montreal (UQAM), 201 President-Kennedy Av., Montréal, QC, H2X 3Y7, Canada.,Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Simon Drouin
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Mariia Samoilenko
- Department of Mathematics, University of Quebec at Montreal (UQAM), 201 President-Kennedy Av., Montréal, QC, H2X 3Y7, Canada.,Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Sophia Morel
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Maja Krajinovic
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Caroline Laverdière
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Daniel Sinnett
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada.,Department of Pediatrics, Faculty of Medicine, University of Montreal, Montréal, QC, H3T 1C5, Canada
| | - Emile Levy
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Valérie Marcil
- Division of Hematology-Oncology, Research Center, Sainte-Justine University Health Center, 3175 Chemin de la Côte-Sainte-Catherine, Montréal, QC, H3T 1C5, Canada
| | - Geneviève Lefebvre
- Department of Mathematics, University of Quebec at Montreal (UQAM), 201 President-Kennedy Av., Montréal, QC, H2X 3Y7, Canada. .,Faculty of Pharmacy, University of Montreal, Montréal, QC, H3T 1J4, Canada. .,Research Center, Centre hospitalier de l'Université de Montréal, Montréal, Canada.
| |
Collapse
|
95
|
Pasvolsky O, Morelli O, Rozovski U, Vaturi M, Wolach O, Amitai I, Vaxman I, Ratzon R, Yeshurun M, Kornowski R, Iakobishvilli Z, Raanani P. Anthracycline-Induced Cardiotoxicity in Acute Myeloid Leukemia Patients Who Undergo Allogeneic Hematopoietic Stem Cell Transplantation. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2019; 19:e343-e348. [PMID: 30948329 DOI: 10.1016/j.clml.2019.03.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Revised: 02/14/2019] [Accepted: 03/01/2019] [Indexed: 12/22/2022]
Abstract
INTRODUCTION There is paucity of data regarding the cardiotoxic effects of anthracycline treatment in the context of acute myeloid leukemia (AML) patients who undergo allogeneic hematopoietic stem cell transplantation (HSCT). Even a transient decrease in cardiac function might affect transplantation outcome. PATIENTS AND METHODS We reviewed the clinical records and echocardiography examinations of 78 patients with AML who received induction therapy and underwent HSCT. RESULTS Twenty-two patients (28%) received daunorubicin at a dose of 90 mg/m2 per day and 53 patients (68%) received 60 mg/m2 per day or an equivalent dose of idarubicin. In 14 patients (18%) the postinduction ejection fraction declined by at least 10%. This change was temporary in 6 patients and longstanding in the remainder. Patients who developed systolic dysfunction had inferior overall survival (13 months compared with 27 months; P = .013). Patients whose diastolic function deteriorated had improved survival outcome (38 months compared with 17 months; P = .048). CONCLUSION Although even transient reduction in systolic function might compromise survival outcome, diastolic dysfunction predicts improved survival in patients with AML who undergo HSCT.
Collapse
Affiliation(s)
- Oren Pasvolsky
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Olga Morelli
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Cardiology, Rabin Medical Center, Petah-Tikva, Israel
| | - Uri Rozovski
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mordehay Vaturi
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Cardiology, Rabin Medical Center, Petah-Tikva, Israel
| | - Ofir Wolach
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Irina Amitai
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Iuliana Vaxman
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Roy Ratzon
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Geha Mental Health Center, Petah-Tikva, Israel
| | - Moshe Yeshurun
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ran Kornowski
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Zaza Iakobishvilli
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Pia Raanani
- Institute of Hematology, Davidoff Cancer Center, Rabin Medical Center, Petah-Tikva, Israel; Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
96
|
Rodrigues PG, Miranda-Silva D, Costa SM, Barros C, Hamdani N, Moura C, Mendes MJ, Sousa-Mendes C, Trindade F, Fontoura D, Vitorino R, Linke WA, Leite-Moreira AF, Falcão-Pires I. Early myocardial changes induced by doxorubicin in the nonfailing dilated ventricle. Am J Physiol Heart Circ Physiol 2019; 316:H459-H475. [DOI: 10.1152/ajpheart.00401.2018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Several studies have demonstrated that administration of doxorubicin (DOXO) results in cardiotoxicity, which eventually progresses to dilated cardiomyopathy. The present work aimed to evaluate the early myocardial changes of DOXO-induced cardiotoxicity. Male New Zealand White rabbits were injected intravenously with DOXO twice weekly for 8 wk [DOXO-induced heart failure (DOXO-HF)] or with an equivolumetric dose of saline (control). Echocardiographic evaluation was performed, and myocardial samples were collected to evaluate myocardial cellular and molecular modifications. The DOXO-HF group presented cardiac hypertrophy and higher left ventricular cavity diameters, showing a dilated phenotype but preserved ejection fraction. Concerning cardiomyocyte function, the DOXO-HF group presented a trend toward increased active tension without significant differences in passive tension. The myocardial GSSG-to-GSH ratio and interstitial fibrosis were increased and Bax-to- Bcl-2 ratio presented a trend toward an increase, suggesting the activation of apoptosis signaling pathways. The macromolecule titin shifted toward the more compliant isoform (N2BA), whereas the stiffer one (N2B) was shown to be hypophosphorylated. Differential protein analysis from the aggregate-enriched fraction through gel liquid chromatography-tandem mass spectrometry revealed an increase in the histidine-rich glycoprotein fragment in DOXO-HF animals. This work describes novel and early myocardial effects of DOXO-induced cardiotoxicity. Thus, tracking these changes appears to be of extreme relevance for the early detection of cardiac damage (as soon as ventricular dilation becomes evident) before irreversible cardiac function deterioration occurs (reduced ejection fraction). Moreover, it allows for the adjustment of the therapeutic approach and thus the prevention of cardiomyopathy progression. NEW & NOTEWORTHY Identification of early myocardial effects of doxorubicin in the heart is essential to hinder the development of cardiac complications and adjust the therapeutic approach. This study describes doxorubicin-induced cellular and molecular modifications before the onset of dilated cardiomyopathy. Myocardial samples from doxorubicin-treated rabbits showed a tendency for higher cardiomyocyte active tension, titin isoform shift from N2B to N2BA, hypophosphorylation of N2B, increased apoptotic genes, left ventricular interstitial fibrosis, and increased aggregation of histidine-rich glycoprotein.
Collapse
Affiliation(s)
- Patricia G. Rodrigues
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Daniela Miranda-Silva
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Sofia M. Costa
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Carla Barros
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Nazha Hamdani
- Department of Systems Physiology, Ruhr University, Bochum, Germany
| | - Cláudia Moura
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Maria J. Mendes
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Cláudia Sousa-Mendes
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Fábio Trindade
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
- Department of Medical Sciences, Institute of Biomedicine, University of Aveiro, Aveiro, Portugal
| | - Dulce Fontoura
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| | - Rui Vitorino
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
- Department of Medical Sciences, Institute of Biomedicine, University of Aveiro, Aveiro, Portugal
| | - Wolfgang A. Linke
- Institute of Physiology II, University of Muenster, Muenster, Germany
| | - Adelino F. Leite-Moreira
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
- Department of Cardiothoracic Surgery, São João Hospital Centre, Porto, Portugal
| | - Inês Falcão-Pires
- Department of Surgery and Physiology, Faculty of Medicine, Unidade de Investigação Cardiovascular, Universidade do Porto, Porto, Portugal
| |
Collapse
|
97
|
Suppression of chemotherapy-induced cytokine/lipid mediator surge and ovarian cancer by a dual COX-2/sEH inhibitor. Proc Natl Acad Sci U S A 2019; 116:1698-1703. [PMID: 30647111 DOI: 10.1073/pnas.1803999116] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Although chemotherapy is a conventional cancer treatment, it may induce a protumorigenic microenvironment by triggering the release of proinflammatory mediators. In this study, we demonstrate that ovarian tumor cell debris generated by first-line platinum- and taxane-based chemotherapy accelerates tumor progression by stimulating a macrophage-derived "surge" of proinflammatory cytokines and bioactive lipids. Thus, targeting a single inflammatory mediator or pathway is unlikely to prevent therapy-induced tumor progression. Here, we show that combined pharmacological abrogation of the cyclooxygenase-2 (COX-2) and soluble epoxide hydrolase (sEH) pathways prevented the debris-induced surge of both cytokines and lipid mediators by macrophages. In animal models, the dual COX-2/sEH inhibitor PTUPB delayed the onset of debris-stimulated ovarian tumor growth and ascites leading to sustained survival over 120 days postinjection. Therefore, dual inhibition of COX-2/sEH may be an approach to suppress debris-stimulated ovarian tumor growth by preventing the therapy-induced surge of cytokines and lipid mediators.
Collapse
|
98
|
Nebigil CG, Désaubry L. Updates in Anthracycline-Mediated Cardiotoxicity. Front Pharmacol 2018; 9:1262. [PMID: 30483123 PMCID: PMC6240592 DOI: 10.3389/fphar.2018.01262] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 10/16/2018] [Indexed: 12/14/2022] Open
Abstract
Cardiotoxicity is one of the main adverse effects of chemotheraphy, affecting the completion of cancer therapies and the short- and long-term quality of life. Anthracyclines are currently used to treat many cancers, including the various forms of leukemia, lymphoma, melanoma, uterine, breast, and gastric cancers. World Health Organization registered anthracyclines in the list of essential medicines. However, anthracyclines display a major cardiotoxicity that can ultimately culminate in congestive heart failure. Taking into account the growing rate of cancer survivorship, the clinical significance of anthracycline cardiotoxicity is an emerging medical issue. In this review, we focus on the key progenitor cells and cardiac cells (cardiomyocytes, fibroblasts, and vascular cells), focusing on the signaling pathways involved in cellular damage, and the clinical biomarkers in anthracycline-mediated cardiotoxicity.
Collapse
Affiliation(s)
- Canan G. Nebigil
- CNRS, Laboratory of Biomolecules, UMR 7203, Sorbonne University, Paris, France
| | | |
Collapse
|
99
|
Barel G, Herwig R. Network and Pathway Analysis of Toxicogenomics Data. Front Genet 2018; 9:484. [PMID: 30405693 PMCID: PMC6204403 DOI: 10.3389/fgene.2018.00484] [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] [Received: 06/19/2018] [Accepted: 09/28/2018] [Indexed: 12/20/2022] Open
Abstract
Toxicogenomics is the study of the molecular effects of chemical, biological and physical agents in biological systems, with the aim of elucidating toxicological mechanisms, building predictive models and improving diagnostics. The vast majority of toxicogenomics data has been generated at the transcriptome level, including RNA-seq and microarrays, and large quantities of drug-treatment data have been made publicly available through databases and repositories. Besides the identification of differentially expressed genes (DEGs) from case-control studies or drug treatment time series studies, bioinformatics methods have emerged that infer gene expression data at the molecular network and pathway level in order to reveal mechanistic information. In this work we describe different resources and tools that have been developed by us and others that relate gene expression measurements with known pathway information such as over-representation and gene set enrichment analyses. Furthermore, we highlight approaches that integrate gene expression data with molecular interaction networks in order to derive network modules related to drug toxicity. We describe the two main parts of the approach, i.e., the construction of a suitable molecular interaction network as well as the conduction of network propagation of the experimental data through the interaction network. In all cases we apply methods and tools to publicly available rat in vivo data on anthracyclines, an important class of anti-cancer drugs that are known to induce severe cardiotoxicity in patients. We report the results and functional implications achieved for four anthracyclines (doxorubicin, epirubicin, idarubicin, and daunorubicin) and compare the information content inherent in the different computational approaches.
Collapse
Affiliation(s)
| | - Ralf Herwig
- Department Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Berlin, Germany
| |
Collapse
|
100
|
Interleukin-1 Beta-A Friend or Foe in Malignancies? Int J Mol Sci 2018; 19:ijms19082155. [PMID: 30042333 PMCID: PMC6121377 DOI: 10.3390/ijms19082155] [Citation(s) in RCA: 301] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/14/2018] [Accepted: 07/19/2018] [Indexed: 12/19/2022] Open
Abstract
Interleukin-1 beta (IL-1β) is induced by inflammatory signals in a broad number of immune cell types. IL-1β (and IL-18) are the only cytokines which are processed by caspase-1 after inflammasome-mediated activation. This review aims to summarize current knowledge about parameters of regulation of IL-1β expression and its multi-facetted role in pathophysiological conditions. IL-1 signaling activates innate immune cells including antigen presenting cells, and drives polarization of CD4+ T cells towards T helper type (Th) 1 and Th17 cells. Therefore, IL-1β has been attributed a largely beneficial role in resolving acute inflammations, and by initiating adaptive anti-tumor responses. However, IL-1β generated in the course of chronic inflammation supports tumor development. Furthermore, IL-1β generated within the tumor microenvironment predominantly by tumor-infiltrating macrophages promotes tumor growth and metastasis via different mechanisms. These include the expression of IL-1 targets which promote neoangiogenesis and of soluble mediators in cancer-associated fibroblasts that evoke antiapoptotic signaling in tumor cells. Moreover, IL-1 promotes the propagation of myeloid-derived suppressor cells. Using genetic mouse models as well as agents for pharmacological inhibition of IL-1 signaling therapeutically applied for treatment of IL-1 associated autoimmune diseases indicate that IL-1β is a driver of tumor induction and development.
Collapse
|