101
|
Hajek E, Krebs F, Bent R, Haas K, Bast A, Steinmetz I, Tuettenberg A, Grabbe S, Bros M. BRAF inhibitors stimulate inflammasome activation and interleukin 1 beta production in dendritic cells. Oncotarget 2018; 9:28294-28308. [PMID: 29983861 PMCID: PMC6033361 DOI: 10.18632/oncotarget.25511] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 05/13/2018] [Indexed: 12/21/2022] Open
Abstract
Melanoma is the most dangerous form of skin cancer with a growing incidence over the last decades. Fourty percent of all melanomas harbor a mutation in the signaling adaptor BRAF (V600E) that results in ERK hyperactivity as an oncogenic driver. In these cases, treatment with the BRAFV600E inhibitors Vemurafenib (VEM) or Dabrafenib (DAB) coapplied with the MEK1/2 inhibitors Cobimetinib (COB) or Trametinib (TRA) can result in long-term suppression of tumor growth. Besides direct suppression of ERK activity, these inhibitors have been reported to also modulate tumor immune responses, and exert pro-inflammatory side effects such as fever and rash in some patients. Here we asked for potential effects of BRAFV600E inhibitors on dendritic cells (DC) which are essential for the induction of adaptive anti-tumor responses. Both splenic and bone marrow-derived (BM) mouse dendritic cells (DC) up-regulated costimulator expression (CD80, CD86) in response to DAB but not VEM treatment. Moreover, DAB and to lesser extent VEM enhanced IL-1β (interleukin 1 beta) release by splenic DC, and by LPS-stimulated BMDC. We demonstrate that DAB and VEM activated the NLRC4/Caspase-1 inflammasome. At high concentration, DAB also induced inflammasome activation independent of Caspase-1. TRA and COB elevated MHCII expression on BMDC, and modulated the LPS-induced cytokine pattern. Immunomodulatory activity of DAB and VEM was also observed in human monocyte-derived DC, and DAB induced IL-1β in human primary DC. Altogether, our study shows that BRAFV600E inhibitors upregulate IL-1β release by mouse and human DC which may affect the DC-mediated course of anti-tumor immune responses.
Collapse
Affiliation(s)
- Eva Hajek
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Franziska Krebs
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Rebekka Bent
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Katharina Haas
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Antje Bast
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Ivo Steinmetz
- Friedrich Loeffler Institute of Medical Microbiology, University Medicine Greifswald, Greifswald, Germany
| | - Andrea Tuettenberg
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
102
|
Santos JC, Pyter LM. Neuroimmunology of Behavioral Comorbidities Associated With Cancer and Cancer Treatments. Front Immunol 2018; 9:1195. [PMID: 29930550 PMCID: PMC6001368 DOI: 10.3389/fimmu.2018.01195] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/14/2018] [Indexed: 12/27/2022] Open
Abstract
Behavioral comorbidities (depression, anxiety, fatigue, cognitive disturbances, and neuropathic pain) are prevalent in cancer patients and survivors. These mental and neurological health issues reduce quality-of-life, which is a significant societal concern given the increasing rates of long-term survival after various cancers. Hypothesized causes of behavioral comorbidities with cancer include tumor biology, stress associated with the cancer experience, and cancer treatments. A relatively recent leading mechanism by which these causes contribute to changes in neurobiology that underlie behavior is inflammation. Indeed, both basic and clinical research indicates that peripheral inflammation leads to central inflammation and behavioral changes in other illness contexts. Given the limitations of assessing neuroimmunology in clinical populations, this review primarily synthesizes evidence of neuroimmune and neuroinflammatory changes due to two components of cancer (tumor biology and cancer treatments) that are associated with altered affective-like or cognitive behaviors in rodents. Specifically, alterations in microglia, neuroinflammation, and immune trafficking to the brain are compiled in models of tumors, chemotherapy, and/or radiation. Evidence-based neuronal mechanisms by which these neuroimmune changes may lead to changes in behavior are proposed. Finally, converging evidence in clinical cancer populations is discussed.
Collapse
Affiliation(s)
- Jessica C Santos
- Department of Basic and Applied Immunology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paulo, Brazil
| | - Leah M Pyter
- Departments of Psychiatry and Behavioral Health and Neuroscience, The Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
103
|
Venancio VP, Almeida MR, Antunes LMG. Cocoplum ( Chrysobalanus icaco L.) decreases doxorubicin-induced DNA damage and downregulates Gadd45a , Il-1β , and Tnf-α in vivo. Food Res Int 2018; 105:996-1002. [DOI: 10.1016/j.foodres.2017.12.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/01/2017] [Accepted: 12/02/2017] [Indexed: 01/03/2023]
|
104
|
Sonowal H, Pal P, Shukla K, Saxena A, Srivastava SK, Ramana KV. Aldose reductase inhibitor, fidarestat prevents doxorubicin-induced endothelial cell death and dysfunction. Biochem Pharmacol 2018; 150:181-190. [PMID: 29458045 DOI: 10.1016/j.bcp.2018.02.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/14/2018] [Indexed: 12/14/2022]
Abstract
Despite doxorubicin (Dox) being one of the most widely used chemotherapy agents for breast, blood and lung cancers, its use in colon cancer is limited due to increased drug resistance and severe cardiotoxic side effects that increase mortality associated with its use at high doses. Therefore, better adjuvant therapies are warranted to improve the chemotherapeutic efficacy and to decrease cardiotoxicity. We have recently shown that aldose reductase inhibitor, fidarestat, increases the Dox-induced colon cancer cell death and reduces cardiomyopathy. However, the efficacy of fidarestat in the prevention of Dox-induced endothelial dysfunction, a pathological event critical to cardiovascular complications, is not known. Here, we have examined the effect of fidarestat on Dox-induced endothelial cell toxicity and dysfunction in vitro and in vivo. Incubation of human umbilical vein endothelial cells (HUVECs) with Dox significantly increased the endothelial cell death, and pre-treatment of fidarestat prevented it. Further, fidarestat prevented the Dox-induced oxidative stress, formation of reactive oxygen species (ROS) and activation of Caspase-3 in HUVECs. Fidarestat also prevented Dox-induced monocyte adhesion to HUVECs and expression of ICAM-1 and VCAM-1. Fidarestat pre-treatment to HUVECs restored the Dox-induced decrease in the Nitric Oxide (NO)-levels and eNOS expression. Treatment of HUVECs with Dox caused a significant increase in the activation of NF-κB and expression of various inflammatory cytokines and chemokines which were prevented by fidarestat pre-treatment. Most importantly, fidarestat prevented the Dox-induced mouse cardiac cell hypertrophy and expression of eNOS, iNOS, and 3-Nitrotyrosine in the aorta tissues. Further, fidarestat blunted the Dox-induced expression of various inflammatory cytokines and chemokines in vivo. Thus, our results suggest that by preventing Dox-induced endothelial cytotoxicity and dysfunction, AR inhibitors could avert cardiotoxicity associated with anthracycline chemotherapy.
Collapse
Affiliation(s)
- Himangshu Sonowal
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Pabitra Pal
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kirtikar Shukla
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ashish Saxena
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Satish K Srivastava
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kota V Ramana
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
105
|
Shaker RA, Abboud SH, Assad HC, Hadi N. Enoxaparin attenuates doxorubicin induced cardiotoxicity in rats via interfering with oxidative stress, inflammation and apoptosis. BMC Pharmacol Toxicol 2018; 19:3. [PMID: 29321061 PMCID: PMC5763526 DOI: 10.1186/s40360-017-0184-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 11/23/2017] [Indexed: 02/06/2023] Open
Abstract
Background Doxorubicin (DOX) is commonly used in the treatment of many types of cancers but its cardiotoxicity is limiting its clinical use. Beyond its anticoagulant action, enoxaparin (ENX), a low molecular weight heparin, has been shown to exert multiple pharmacological actions including antioxidant, anti-inflammatory and antiapoptotic effects. Therefore, the current study aimed to assess if ENX could ameliorate cardiotoxicity induced by DOX. Methods Twenty-one adult male Wistar albino rats were randomly allocated into three groups (n = 7 each) of control, receiving 0.9% saline (i.p.), DOX, receiving 2.5 mg/kg of DOX (i.p.) thrice weekly; and DOX + ENX, receiving ENX (250 IU/kg/day i.p.) and a DOX dose equivalent to that of the DOX only group. Results DOX-induced cardiotoxicity was indicated by marked increases in cardiac troponin I (cTnI) and severe histological lesions, which significantly correlated with cardiotoxicity, oxidative stress, inflammation and apoptosis markers, compared to controls. DOX group also showed elevations in malondialdehyde (MDA), a marker of oxidative stress, and reductions in total antioxidant capacity (TAC). Cardiac inflammatory markers including tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1β) and caspase-3, an apoptotic marker, were also elevated in the DOX group. DOX, however, did not significantly alter brain natriuretic peptide (BNP) levels. ENX significantly attenuated, but not completely reversed, DOX-induced cardiotoxicity through lowering cTnI and improving cardiomyopathy histopathological scores as compared to the DOX group. ENX also decreased MDA, increased TAC of rats’ heart to levels relatively comparable to control. Significant reductions in TNF-α, IL-1β and caspase-3 were also observed following ENX treatment relative to the DOX only group. Conclusions Collectively, these results describe a cardioprotective effect for ENX against DOX-induced cardiotoxicity which is likely facilitated via suppression of oxidative stress, inflammation and apoptosis.
Collapse
Affiliation(s)
- Reem Ali Shaker
- Najaf Health Directorate, Ministry of Health, Najaf Governorate, Iraq
| | | | - Hayder Chasib Assad
- Department of Clinical Pharmacy and Therapeutics, Faculty of Pharmacy, University of Kufa, Najaf Governorate, Iraq.
| | - Najah Hadi
- Faculty of Medicine, University of Kufa, Najaf Governorate, Iraq
| |
Collapse
|
106
|
Molecular mechanism of doxorubicin-induced cardiomyopathy - An update. Eur J Pharmacol 2017; 818:241-253. [PMID: 29074412 DOI: 10.1016/j.ejphar.2017.10.043] [Citation(s) in RCA: 394] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 10/11/2017] [Accepted: 10/20/2017] [Indexed: 12/27/2022]
Abstract
Doxorubicin is utilized for anti-neoplastic treatment for several decades. The utility of this drug is limited due to its side effects. Generally, doxorubicin toxicity is originated from the myocardium and then other organs are also ruined. The mechanism of doxorubicin is intercalated with the DNA and inhibits topoisomerase 2. There are various signalling mechanisms involved in doxorubicin cardiotoxicity. First and foremost, the doxorubicin-induced cardiotoxicity is due to oxidative stress. Cardiac mitochondrial damage is supposed after few hours following the revelation of doxorubicin. This has led important new uses for the mechanism of doxorubicin-induced cardiotoxicity and novel avenues of investigation to determine better pharmacotherapies and interventions for the impediment of cardiotoxicity. The idea of this review is to bring up to date the recent findings of the mechanism of doxorubicin cardiomyopathies such as calcium dysregulation, endoplasmic reticulum stress, impairment of progenitor cells, activation of immune, ubiquitous system and some other parameters.
Collapse
|
107
|
Cardiac Nonmyocyte Cell Functions and Crosstalks in Response to Cardiotoxic Drugs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1089359. [PMID: 29201269 PMCID: PMC5671742 DOI: 10.1155/2017/1089359] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/24/2017] [Indexed: 01/06/2023]
Abstract
The discovery of the molecular mechanisms involved in the cardiac responses to anticancer drugs represents the current goal of cardio-oncology research. The oxidative stress has a pivotal role in cardiotoxic responses, affecting the function of all types of cardiac cells, and their functional crosstalks. Generally, cardiomyocytes are the main target of research studies on cardiotoxicity, but recently the contribution of the other nonmyocyte cardiac cells is becoming of growing interest. This review deals with the role of oxidative stress, induced by anticancer drugs, in cardiac nonmyocyte cells (fibroblasts, vascular cells, and immune cells). The alterations of functional interplays among these cardiac cells are discussed, as well. These interesting recent findings increase the knowledge about cardiotoxicity and suggest new molecular targets for both diagnosis and therapy.
Collapse
|
108
|
El-Sayed ESM, Mansour AM, El-Sawy WS. Protective effect of proanthocyanidins against doxorubicin-induced nephrotoxicity in rats. J Biochem Mol Toxicol 2017; 31. [DOI: 10.1002/jbt.21965] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Revised: 07/03/2017] [Accepted: 07/11/2017] [Indexed: 02/05/2023]
Affiliation(s)
- El-Sayed M. El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy; Al-Azhar University; Cairo Egypt
| | - Ahmed M. Mansour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy; Al-Azhar University; Cairo Egypt
| | - Waleed S. El-Sawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy; Al-Azhar University; Assiut Egypt
| |
Collapse
|
109
|
El-Sayed ESM, Mansour AM, El-Sawy WS. Alpha lipoic acid prevents doxorubicin-induced nephrotoxicity by mitigation of oxidative stress, inflammation, and apoptosis in rats. J Biochem Mol Toxicol 2017; 31. [DOI: 10.1002/jbt.21940] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 05/16/2017] [Accepted: 05/20/2017] [Indexed: 02/05/2023]
Affiliation(s)
- El-Sayed M. El-Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy; Al-Azhar University; Cairo Egypt
| | - Ahmed M. Mansour
- Department of Pharmacology and Toxicology, Faculty of Pharmacy; Al-Azhar University; Cairo Egypt
| | - Waleed S. El-Sawy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy; Al-Azhar University; Assiut Egypt
| |
Collapse
|
110
|
Borniger JC, Cisse YM, Surbhi, Nelson RJ. Reciprocal Regulation of Circadian Rhythms and Immune Function. CURRENT SLEEP MEDICINE REPORTS 2017. [DOI: 10.1007/s40675-017-0070-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
111
|
Targeting the interleukin-1 pathway in patients with hematological disorders. Blood 2017; 129:3155-3164. [PMID: 28483765 DOI: 10.1182/blood-2016-12-754994] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 04/23/2017] [Indexed: 12/14/2022] Open
Abstract
Interleukin-1α (IL-1α) and IL-1β are potent inflammatory cytokines that activate local and systemic inflammatory processes and are involved in protective immune responses against infections. However, their dysregulated production and signaling can aggravate tissue damage during infection, inflammatory diseases, and chemotherapy-induced intestinal mucositis. Additionally, cytokines of the IL-1 family play an important role in homeostatic as well as "emergency" hematopoiesis and are involved in the pathogenesis of several myeloid and lymphoid hematological malignancies. In the pathogenesis of intestinal mucositis and graft-versus-host disease (GVHD), these cytokines are considered pivotal during the initiation as well as propagation phase, and insights from animal studies suggest that targeting the IL-1 pathway can significantly ameliorate mucositis and GVHD. Moreover, IL-1α and IL-1β might prove to be valuable targets for both prevention and treatment of cancer and cancer therapy-related complications, and the first clinical studies have already been performed in the setting of hematological malignancies. In this review, we will discuss the role of cytokines of the IL-1 family in hematological malignancies, chemotherapy-induced intestinal mucositis, and GVHD, and speculate on possibilities of therapeutically targeting the IL-1 pathway in hematological patients.
Collapse
|
112
|
Prevention of Adriamycin induced cardiotoxicity in rats — A comparative study with subacute angiotensin-converting enzyme inhibitor and nonselective beta blocker therapy. ACTA ACUST UNITED AC 2017. [DOI: 10.1016/j.ijcme.2017.01.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
113
|
Dysregulation of cytokine mediated chemotherapy induced cognitive impairment. Pharmacol Res 2017; 117:267-273. [DOI: 10.1016/j.phrs.2017.01.001] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 01/03/2017] [Indexed: 11/19/2022]
|
114
|
Roerink ME, van der Schaaf ME, Dinarello CA, Knoop H, van der Meer JWM. Interleukin-1 as a mediator of fatigue in disease: a narrative review. J Neuroinflammation 2017; 14:16. [PMID: 28109186 PMCID: PMC5251329 DOI: 10.1186/s12974-017-0796-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/12/2017] [Indexed: 01/17/2023] Open
Abstract
Fatigue is commonly reported in a variety of illnesses, and it has major impact on quality of life. Previously, it was thought that fatigue originates in the skeletal muscles, leading to cessation of activity. However, more recently, it has become clear that the brain is the central regulator of fatigue perception. It has been suggested that pro-inflammatory cytokines, especially interleukin-1 alpha (IL-1α) and interleukin-1 beta (IL-1β), play a prominent role in the development of central fatigue, and several studies have been performed to elucidate the connection between inflammation and these central processes.In this narrative review, mechanisms of action of IL-1 are described, with special attention to its effect on the central nervous system. In addition, we present a summary of studies that (i) investigated the relationship between circulating IL-1α and IL-1β and fatigue severity and/or (ii) evaluated the effect of inhibiting IL-1 on fatigue. We aim to improve the understanding of fatigue in both inflammatory and non-inflammatory illnesses, which could help develop strategies to treat fatigue more effectively.Reviewing the studies that have been performed, it appears that there is a limited value of measuring circulating IL-1. However, inhibiting IL-1 has a positive effect on severe fatigue in most studies that have been conducted.
Collapse
Affiliation(s)
- Megan E Roerink
- Department of Internal Medicine, Radboud University Medical Centre, Geert Grooteplein Zuid 8, 6500HB, Nijmegen, The Netherlands.
| | - Marieke E van der Schaaf
- Expert Centre for Chronic Fatigue, Radboud University Medical Centre, Reinier Postlaan 4, 6525GC, Nijmegen, The Netherlands
| | - Charles A Dinarello
- Department of Internal Medicine, Radboud University Medical Centre, Geert Grooteplein Zuid 8, 6500HB, Nijmegen, The Netherlands.,Department of Medicine, University of Colorado Denver, 12700 E. 19th Avenue Box B168, Aurora, CO, 80045, USA
| | - Hans Knoop
- Expert Centre for Chronic Fatigue, Radboud University Medical Centre, Reinier Postlaan 4, 6525GC, Nijmegen, The Netherlands.,Department of Medical Psychology, Academic Medical Centre (AMC), University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands
| | - Jos W M van der Meer
- Department of Internal Medicine, Radboud University Medical Centre, Geert Grooteplein Zuid 8, 6500HB, Nijmegen, The Netherlands
| |
Collapse
|
115
|
Lynch Kelly D, Dickinson K, Hsiao CP, Lukkahatai N, Gonzalez-Marrero V, McCabe M, Saligan LN. Biological Basis for the Clustering of Symptoms. Semin Oncol Nurs 2016; 32:351-360. [PMID: 27776832 DOI: 10.1016/j.soncn.2016.08.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVES Identification of biologic pathways of symptom clusters is necessary to develop precision therapies for distressing symptoms. This review examined extant literature evaluating relationships between biomarkers and symptom clusters in cancer survivors. DATA SOURCES PubMed, CINAHL, Web of Science and Cochrane Library were searched using terms "biological markers" or "biomarkers" and "symptom cluster" or "symptom complex" or "multiple symptoms." CONCLUSION Biomarkers related to inflammation (eg, cytokines) were the most studied and showed the most significant relationships with clusters of symptoms. This review suggests that clustering of symptoms related to cancer or cancer therapy is linked to immune/inflammatory pathways. IMPLICATIONS FOR NURSING PRACTICE Understanding the etiology of symptom clusters may guide future nursing interventions for symptom management.
Collapse
|
116
|
Cuyàs E, Martin-Castillo B, Corominas-Faja B, Massaguer A, Bosch-Barrera J, Menendez JA. Anti-protozoal and anti-bacterial antibiotics that inhibit protein synthesis kill cancer subtypes enriched for stem cell-like properties. Cell Cycle 2016; 14:3527-32. [PMID: 25970790 PMCID: PMC4825717 DOI: 10.1080/15384101.2015.1044173] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Key players in translational regulation such as ribosomes might represent powerful, but hitherto largely unexplored, targets to eliminate drug-refractory cancer stem cells (CSCs). A recent study by the Lisanti group has documented how puromycin, an old antibiotic derived from Streptomyces alboniger that inhibits ribosomal protein translation, can efficiently suppress CSC states in tumorspheres and monolayer cultures. We have used a closely related approach based on Biolog Phenotype Microarrays (PM), which contain tens of lyophilized antimicrobial drugs, to assess the chemosensitivity profiles of breast cancer cell lines enriched for stem cell-like properties. Antibiotics directly targeting active sites of the ribosome including emetine, puromycin and cycloheximide, inhibitors of ribosome biogenesis such as dactinomycin, ribotoxic stress agents such as daunorubicin, and indirect inhibitors of protein synthesis such as acriflavine, had the largest cytotoxic impact against claudin-low and basal-like breast cancer cells. Thus, biologically aggressive, treatment-resistant breast cancer subtypes enriched for stem cell-like properties exhibit exacerbated chemosensitivities to anti-protozoal and anti-bacterial antibiotics targeting protein synthesis. These results suggest that old/existing microbicides might be repurposed not only as new cancer therapeutics, but also might provide the tools and molecular understanding needed to develop second-generation inhibitors of ribosomal translation to eradicate CSC traits in tumor tissues.
Collapse
Affiliation(s)
- Elisabet Cuyàs
- a Metabolism & Cancer Group; Translational Research Laboratory; Catalan Institute of Oncology (ICO) ; Girona; Catalonia , Spain.,b Girona Biomedical Research Institute (IDIBGI) ; Girona, Catalonia , Spain
| | - Begoña Martin-Castillo
- b Girona Biomedical Research Institute (IDIBGI) ; Girona, Catalonia , Spain.,c Unit of Clinical Research ; Catalan Institute of Oncology (ICO) ; Girona, Catalonia , Spain
| | - Bruna Corominas-Faja
- a Metabolism & Cancer Group; Translational Research Laboratory; Catalan Institute of Oncology (ICO) ; Girona; Catalonia , Spain.,b Girona Biomedical Research Institute (IDIBGI) ; Girona, Catalonia , Spain
| | - Anna Massaguer
- d Biochemistry and Molecular Biology Unit ; Department of Biology; University of Girona ; Girona, Catalonia , Spain
| | - Joaquim Bosch-Barrera
- b Girona Biomedical Research Institute (IDIBGI) ; Girona, Catalonia , Spain.,e Medical Oncology Department ; Catalan Institute of Oncology (ICO) ; Girona, Catalonia , Spain
| | - Javier A Menendez
- a Metabolism & Cancer Group; Translational Research Laboratory; Catalan Institute of Oncology (ICO) ; Girona; Catalonia , Spain.,b Girona Biomedical Research Institute (IDIBGI) ; Girona, Catalonia , Spain
| |
Collapse
|
117
|
Vince JE, Silke J. The intersection of cell death and inflammasome activation. Cell Mol Life Sci 2016; 73:2349-67. [PMID: 27066895 PMCID: PMC11108284 DOI: 10.1007/s00018-016-2205-2] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 02/07/2023]
Abstract
Inflammasomes sense cellular danger to activate the cysteine-aspartic protease caspase-1, which processes precursor interleukin-1β (IL-1β) and IL-18 into their mature bioactive fragments. In addition, activated caspase-1 or the related inflammatory caspase, caspase-11, can cleave gasdermin D to induce a lytic cell death, termed pyroptosis. The intertwining of IL-1β activation and cell death is further highlighted by research showing that the extrinsic apoptotic caspase, caspase-8, may, like caspase-1, directly process IL-1β, activate the NLRP3 inflammasome itself, or bind to inflammasome complexes to induce apoptotic cell death. Similarly, RIPK3- and MLKL-dependent necroptotic signaling can activate the NLRP3 inflammasome to drive IL-1β inflammatory responses in vivo. Here, we review the mechanisms by which cell death signaling activates inflammasomes to initiate IL-1β-driven inflammation, and highlight the clinical relevance of these findings to heritable autoinflammatory diseases. We also discuss whether the act of cell death can be separated from IL-1β secretion and evaluate studies suggesting that several cell death regulatory proteins can directly interact with, and modulate the function of, inflammasome and IL-1β containing protein complexes.
Collapse
Affiliation(s)
- James E Vince
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3050, Australia.
| | - John Silke
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, VIC, 3050, Australia
| |
Collapse
|
118
|
NLRP3 Deficiency Reduces Macrophage Interleukin-10 Production and Enhances the Susceptibility to Doxorubicin-induced Cardiotoxicity. Sci Rep 2016; 6:26489. [PMID: 27225830 PMCID: PMC4880937 DOI: 10.1038/srep26489] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/04/2016] [Indexed: 01/04/2023] Open
Abstract
NLRP3 inflammasomes recognize non-microbial danger signals and induce release of proinflammatory cytokine interleukin (IL)-1β, leading to sterile inflammation in cardiovascular disease. Because sterile inflammation is involved in doxorubicin (Dox)-induced cardiotoxicity, we investigated the role of NLRP3 inflammasomes in Dox-induced cardiotoxicity. Cardiac dysfunction and injury were induced by low-dose Dox (15 mg/kg) administration in NLRP3-deficient (NLRP3−/−) mice but not in wild-type (WT) and IL-1β−/− mice, indicating that NLRP3 deficiency enhanced the susceptibility to Dox-induced cardiotoxicity independent of IL-1β. Although the hearts of WT and NLRP3−/− mice showed no significant difference in inflammatory cell infiltration, macrophages were the predominant inflammatory cells in the hearts, and cardiac IL-10 production was decreased in Dox-treated NLRP3−/− mice. Bone marrow transplantation experiments showed that bone marrow-derived cells contributed to the exacerbation of Dox-induced cardiotoxicity in NLRP3−/− mice. In vitro experiments revealed that NLRP3 deficiency decreased IL-10 production in macrophages. Furthermore, adeno-associated virus-mediated IL-10 overexpression restored the exacerbation of cardiotoxicity in the NLRP3−/− mice. These results demonstrated that NLRP3 regulates macrophage IL-10 production and contributes to the pathophysiology of Dox-induced cardiotoxicity, which is independent of IL-1β. Our findings identify a novel role of NLRP3 and provided new insights into the mechanisms underlying Dox-induced cardiotoxicity.
Collapse
|
119
|
Marchetti C, Toldo S, Chojnacki J, Mezzaroma E, Liu K, Salloum FN, Nordio A, Carbone S, Mauro AG, Das A, Zalavadia AA, Halquist MS, Federici M, Van Tassell BW, Zhang S, Abbate A. Pharmacologic Inhibition of the NLRP3 Inflammasome Preserves Cardiac Function After Ischemic and Nonischemic Injury in the Mouse. J Cardiovasc Pharmacol 2016; 66:1-8. [PMID: 25915511 DOI: 10.1097/fjc.0000000000000247] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Sterile inflammation resulting from myocardial injury activates the NLRP3 inflammasome and amplifies the inflammatory response mediating further damage. METHODS We used 2 experimental models of ischemic injury (acute myocardial infarction [AMI] with and without reperfusion) and a model of nonischemic injury due to doxorubicin 10 mg/kg to determine whether the NLRP3 inflammasome preserved cardiac function after injury. RESULTS Treatment with the NLRP3 inflammasome inhibitor in the reperfused AMI model caused a significant reduction in infarct size measured at pathology or as serum cardiac troponin I level (-56% and -82%, respectively, both P < 0.001) and preserved left ventricular fractional shortening (LVFS, 31 ± 2 vs. vehicle 26% ± 1%, P = 0.003). In the non-reperfused AMI model, treatment with the NLRP3 inhibitor significantly limited LV systolic dysfunction at 7 days (LVFS of 20 ± 2 vs. 14% ± 1%, P = 0.002), without a significant effect on infarct size. In the doxorubicin model, a significant increase in myocardial interstitial fibrosis and a decline in systolic function were seen in vehicle-treated mice, whereas treatment with the NLRP3 inhibitor significantly reduced fibrosis (-80%, P = 0.001) and preserved systolic function (LVFS 35 ± 2 vs. vehicle 27% ± 2%, P = 0.017). CONCLUSIONS Pharmacological inhibition of the NLRP3 inflammasome limits cell death and LV systolic dysfunction after ischemic and nonischemic injury in the mouse.
Collapse
Affiliation(s)
- Carlo Marchetti
- *VCU Pauley Heart Center, Virginia Commonwealth University, Richmond, VA; †Victoria Johnson Research Laboratories, Richmond, VA; ‡Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Departments of §Medicinal Chemistry; ‖Pharmacotherapy and Outcome Studies, and ¶Pharmaceutics Virginia Commonwealth University, Richmond, VA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
120
|
Docosahexaenoic Acid Attenuates Doxorubicin-induced Cytotoxicity and Inflammation by Suppressing NF-κB/iNOS/NO Signaling Pathway Activation in H9C2 Cardiac Cells. J Cardiovasc Pharmacol 2016; 67:283-9. [DOI: 10.1097/fjc.0000000000000350] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
121
|
Lee EM, Yee D, Busfield SJ, McManus JF, Cummings N, Vairo G, Wei A, Ramshaw HS, Powell JA, Lopez AF, Lewis ID, McCall MN, Lock RB. Efficacy of an Fc-modified anti-CD123 antibody (CSL362) combined with chemotherapy in xenograft models of acute myelogenous leukemia in immunodeficient mice. Haematologica 2016; 100:914-26. [PMID: 26130514 DOI: 10.3324/haematol.2014.113092] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The prognosis of older patients with acute myelogenous leukemia is generally poor. The interleukin-3 receptor α-chain (CD123) is highly expressed on the surface of acute leukemia cells compared with normal hematopoietic stem cells. CSL362 is a fully humanized, CD123-neutralizing monoclonal antibody containing a modified Fc structure, which enhances human natural killer cell antibody-dependent cell-mediated cytotoxicity. Six continuous acute myelogenous leukemia xenografts established from patient explants and characterized by cell and molecular criteria, produced progressively lethal disease 42-202 days after transplantation. CSL362 alone reduced engraftment of one of four and three of four acute myelogenous leukemia xenografts in the bone marrow and peripheral organs, respectively. A cytarabine and daunorubicin regimen was optimized using this model to identify potentially synergistic interactions with CSL362. Cytarabine/daunorubicin improved the survival of mice engrafted with four of four acute myelogenous leukemia xenografts by 31-41 days. Moreover, CSL362 extended the survival of cytarabine/daunorubicin-treated mice for two of two acute myelogenous leukemia xenografts, while augmentation of natural killer cell-deficient NSG mice with adoptively transferred human natural killer cells improved survival against a single xenograft. Interestingly, this enhanced CSL362 efficacy was lost in the absence of chemotherapy. This study shows that acute myelogenous leukemia xenografts provide a platform for the evaluation of new therapeutics, simulating complex in vivo interactions, and that the in vivo efficacy of CSL362 supports continued clinical development of this drug.
Collapse
Affiliation(s)
- Erwin M Lee
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - Dean Yee
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | | | - Julie F McManus
- Department of Microbiology, The Alfred Hospital and Monash University, Melbourne, Australia
| | - Nik Cummings
- Department of Haematology, The Alfred Hospital and Monash University, Melbourne, Australia
| | | | - Andrew Wei
- Department of Haematology, The Alfred Hospital and Monash University, Melbourne, Australia
| | - Hayley S Ramshaw
- The Centre for Cancer Biology, SA Pathology, Adelaide, Australia
| | - Jason A Powell
- The Centre for Cancer Biology, SA Pathology, Adelaide, Australia School of Medicine, University of Adelaide, Adelaide, Australia
| | - Angel F Lopez
- The Centre for Cancer Biology, SA Pathology, Adelaide, Australia
| | - Ian D Lewis
- Division of Haematology and Centre for Cancer Biology, SA Pathology, Adelaide, Australia
| | - Martin N McCall
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| | - Richard B Lock
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, UNSW, Sydney, Australia
| |
Collapse
|
122
|
Sharma M, Tuaine J, McLaren B, Waters DL, Black K, Jones LM, McCormick SPA. Chemotherapy Agents Alter Plasma Lipids in Breast Cancer Patients and Show Differential Effects on Lipid Metabolism Genes in Liver Cells. PLoS One 2016; 11:e0148049. [PMID: 26807857 PMCID: PMC4726544 DOI: 10.1371/journal.pone.0148049] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 01/12/2016] [Indexed: 11/18/2022] Open
Abstract
Cardiovascular complications have emerged as a major concern for cancer patients. Many chemotherapy agents are cardiotoxic and some appear to also alter lipid profiles, although the mechanism for this is unknown. We studied plasma lipid levels in 12 breast cancer patients throughout their chemotherapy. Patients received either four cycles of doxorubicin and cyclophosphamide followed by weekly paclitaxel or three cycles of epirubicin, cyclophosphamide and 5’-fluorouracil followed by three cycles of docetaxel. Patients demonstrated a significant reduction (0.32 mmol/L) in high density lipoprotein cholesterol (HDL-C) and apolipoprotein A1 (apoA1) levels (0.18 g/L) and an elevation in apolipoprotein B (apoB) levels (0.15 g/L) after treatment. Investigation of the individual chemotherapy agents for their effect on genes involved in lipoprotein metabolism in liver cells showed that doxorubicin decreased ATP binding cassette transporter A1 (ABCA1) via a downregulation of the peroxisomal proliferator activated receptor γ (PPARγ) and liver X receptor α (LXRα) transcription factors. In contrast, ABCA1 levels were not affected by cyclophosphamide or paclitaxel. Likewise, apoA1 levels were reduced by doxorubicin and remained unaffected by cyclophosphamide and paclitaxel. Doxorubicin and paclitaxel both increased apoB protein levels and paclitaxel also decreased low density lipoprotein receptor (LDLR) protein levels. These findings correlate with the observed reduction in HDL-C and apoA1 and increase in apoB levels seen in these patients. The unfavourable lipid profiles produced by some chemotherapy agents may be detrimental in the longer term to cancer patients, especially those already at risk of cardiovascular disease (CVD). This knowledge may be useful in tailoring effective follow-up care plans for cancer survivors.
Collapse
Affiliation(s)
- Monika Sharma
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Jo Tuaine
- Southern Blood and Cancer Service, Dunedin Hospital, Dunedin, New Zealand
| | - Blair McLaren
- Southern Blood and Cancer Service, Dunedin Hospital, Dunedin, New Zealand
| | - Debra L. Waters
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | - Katherine Black
- Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | - Lynnette M. Jones
- School of Physical Education, Sport and Exercise Sciences, University of Otago, Dunedin, New Zealand
| | | |
Collapse
|
123
|
Vyas D, Deshpande K, Chaturvedi L, Gieric L, Ching K. Rapid Extensive Recurrence of Triple Negative Breast Cancer: Are Both Therapy and Cancer Biology the Culprit? J Clin Med Res 2015; 8:162-7. [PMID: 26767086 PMCID: PMC4701073 DOI: 10.14740/jocmr2365w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2015] [Indexed: 01/22/2023] Open
Abstract
Triple negative breast cancer (TNBC) comprises 17-20% of all breast cancers and is one of the most common breast cancers. The lack of therapy and failure of existing therapy has been a challenge for clinicians. Doxorubicin (DOX) is the first-line therapy, however, it has significant limitations. Rapid extensive recurrence with metastasis in any cancer has been a challenge for surgeons and medical oncologists. The challenge can be due to failure of therapy, drug resistance, or epigenetic changes. Here, we are discussing a stage I breast cancer patient, operated and treated with appropriate chemotherapy with complete response, which recurred in less than 8 months and metastasized to bone, liver and other organs. We are also presenting lab data of the IL-6 secretions on exposure to DOX in one of the most commonly used TNBC cell lines MDA-MB-231. Breast cancer cell line MDA-MB-231 upon exposure to DOX shows an increase in IL-6 levels more than the already elevated IL-6 levels. This might be a reason for early recurrence. We concluded that patients with TNBC might benefit from a standard DOX treatment regimen with an inflammation-blocking agent.
Collapse
Affiliation(s)
- Dinesh Vyas
- 61391 ODE Surgery, Department of Surgery, Texas Tech University Health Sciences Center, 3601 Fourth Street, Lubbock, TX 79430, USA; These authors contributed equally to this paper
| | - Kaivalya Deshpande
- Michigan State University, College of Human Medicine, 1200 East Michigan Avenue, Suite 655, East Lansing, MI 48824, USA
| | - Lakshmishankar Chaturvedi
- Michigan State University, College of Human Medicine, 1200 East Michigan Avenue, Suite 655, East Lansing, MI 48824, USA
| | - Laput Gieric
- Michigan State University, College of Human Medicine, 1200 East Michigan Avenue, Suite 655, East Lansing, MI 48824, USA; These authors contributed equally to this paper
| | - Karen Ching
- Michigan State University, College of Human Medicine, 1200 East Michigan Avenue, Suite 655, East Lansing, MI 48824, USA
| |
Collapse
|
124
|
Jo EK, Kim JK, Shin DM, Sasakawa C. Molecular mechanisms regulating NLRP3 inflammasome activation. Cell Mol Immunol 2015; 13:148-59. [PMID: 26549800 DOI: 10.1038/cmi.2015.95] [Citation(s) in RCA: 1021] [Impact Index Per Article: 102.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 10/14/2015] [Accepted: 10/14/2015] [Indexed: 02/06/2023] Open
Abstract
Inflammasomes are multi-protein signaling complexes that trigger the activation of inflammatory caspases and the maturation of interleukin-1β. Among various inflammasome complexes, the NLRP3 inflammasome is best characterized and has been linked with various human autoinflammatory and autoimmune diseases. Thus, the NLRP3 inflammasome may be a promising target for anti-inflammatory therapies. In this review, we summarize the current understanding of the mechanisms by which the NLRP3 inflammasome is activated in the cytosol. We also describe the binding partners of NLRP3 inflammasome complexes activating or inhibiting the inflammasome assembly. Our knowledge of the mechanisms regulating NLRP3 inflammasome signaling and how these influence inflammatory responses offers further insight into potential therapeutic strategies to treat inflammatory diseases associated with dysregulation of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 301-747, South Korea.,Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon 301-747, South Korea
| | - Jin Kyung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 301-747, South Korea.,Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon 301-747, South Korea
| | - Dong-Min Shin
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon 301-747, South Korea.,Infection Signaling Network Research Center, Chungnam National University School of Medicine, Daejeon 301-747, South Korea
| | - Chihiro Sasakawa
- Medical Mycology Research Center, Chiba University, Chiba 260-8673, Japan.,Nippon Institute for Biological Science, Tokyo 198-0024, Japan
| |
Collapse
|
125
|
Molinaro M, Ameri P, Marone G, Petretta M, Abete P, Di Lisa F, De Placido S, Bonaduce D, Tocchetti CG. Recent Advances on Pathophysiology, Diagnostic and Therapeutic Insights in Cardiac Dysfunction Induced by Antineoplastic Drugs. BIOMED RESEARCH INTERNATIONAL 2015; 2015:138148. [PMID: 26583088 PMCID: PMC4637019 DOI: 10.1155/2015/138148] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/01/2015] [Indexed: 12/28/2022]
Abstract
Along with the improvement of survival after cancer, cardiotoxicity due to antineoplastic treatments has emerged as a clinically relevant problem. Potential cardiovascular toxicities due to anticancer agents include QT prolongation and arrhythmias, myocardial ischemia and infarction, hypertension and/or thromboembolism, left ventricular (LV) dysfunction, and heart failure (HF). The latter is variable in severity, may be reversible or irreversible, and can occur soon after or as a delayed consequence of anticancer treatments. In the last decade recent advances have emerged in clinical and pathophysiological aspects of LV dysfunction induced by the most widely used anticancer drugs. In particular, early, sensitive markers of cardiac dysfunction that can predict this form of cardiomyopathy before ejection fraction (EF) is reduced are becoming increasingly important, along with novel therapeutic and cardioprotective strategies, in the attempt of protecting cardiooncologic patients from the development of congestive heart failure.
Collapse
Affiliation(s)
- Marilisa Molinaro
- Department of Medicine and Health Sciences, University of Molise, 86100 Campobasso, Italy
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Giancarlo Marone
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | - Mario Petretta
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| | - Pasquale Abete
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy
- National Researches Council, Neuroscience Institute, University of Padova, 35121 Padova, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | - Domenico Bonaduce
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| |
Collapse
|
126
|
Huaiqihuang Granules () reduce proteinuria by enhancing nephrin expression and regulating necrosis factor κB signaling pathway in adriamycin-induced nephropathy. Chin J Integr Med 2015; 23:279-287. [PMID: 26453563 DOI: 10.1007/s11655-015-2293-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Indexed: 01/25/2023]
Abstract
OBJECTIVE To investigate the effects of Huaiqihuang Granules (, HQH), a mixture of Chinese herbs including Trametes robiniophila Murr, Fructus Lycii and Polygonatum sibiricum, on adriamycininduced nephropathy (ADRN) in rats and its underlying mechanisms. METHODS Rats with ADRN were divided into four groups: the sham group, the model group (distilled water), the low-dose HQH-treated (2 g/kg) group, and the high-dose HQH-treated (4 g/kg) group. Body weight and 24-h urinary protein (Upro) were checked every week. After 5-week intervention, at the end of the study, the rats were sacrificed and blood samples were collected for examination of biochemical parameters, including glomerular morphological makers, podocyte shape, cellular apoptosis, expressions of nephrin, inflammatory and apoptosis markers. RESULTS HQH ameliorated the rat's general status, proteinuria, renal morphological appearance and glomerulosclerosis. The decreased expression of nephrin in ADRN rats was increased by HQH, as well as the impaired podocyte foot process fusion. Cytosolic levels of p65 and inhibitor of nuclear factor κBα (IκBα) were decreased in ADRN rats, and recovered by the treatment of HQH. Consistently, the induced expression of tumor necrosis factor α (TNF-α), phosphorylated nuclear factor κB p65 (p-NFκB p65) and IκBα in ADRN were markedly suppressed by HQH. In addition, induction of Bax, cleaved caspase-3 and cytochrome C in ADRN rats were suppressed by HQH, indicating the amelioration of apoptosis. CONCLUSION HQH could ameliorate renal impairments in ADRN rats by increasing nephrin expression, inhibiting NF-κB signaling pathway via the down-regulation of p-NF-κB p65 and p-IκBα, and suppression of glomerular and tubular apoptosis.
Collapse
|
127
|
Borniger JC, Gaudier-Diaz MM, Zhang N, Nelson RJ, DeVries AC. Cytotoxic chemotherapy increases sleep and sleep fragmentation in non-tumor-bearing mice. Brain Behav Immun 2015; 47:218-27. [PMID: 25449581 DOI: 10.1016/j.bbi.2014.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Revised: 10/29/2014] [Accepted: 11/03/2014] [Indexed: 11/27/2022] Open
Abstract
Sleep disruption ranks among the most common complaints of breast cancer patients undergoing chemotherapy. Because of the complex interactions among cancer, treatment regimens, and life-history traits, studies to establish a causal link between chemotherapy and sleep disruption are uncommon. To investigate how chemotherapy acutely influences sleep, adult female c57bl/6 mice were ovariectomized and implanted with wireless biotelemetry units. EEG/EMG biopotentials were collected over the course of 3days pre- and post-injection of 13.5mg/kg doxorubicin and 135mg/kg cyclophosphamide or the vehicle. We predicted that cyclophosphamide+doxorubicin would disrupt sleep and increase central proinflammatory cytokine expression in brain areas that govern vigilance states (i.e., hypothalamus and brainstem). The results largely support these predictions; a single chemotherapy injection increased NREM and REM sleep during subsequent active (dark) phases; this induced sleep was fragmented and of low quality. Mice displayed marked increases in low theta (5-7Hz) to high theta (7-10Hz) ratios following chemotherapy treatment, indicating elevated sleep propensity. The effect was strongest during the first dark phase following injection, but mice displayed disrupted sleep for the entire 3-day duration of post-injection sleep recording. Vigilance state timing was not influenced by treatment, suggesting that acute chemotherapy administration alters sleep homeostasis without altering sleep timing. qPCR analysis revealed that disrupted sleep was accompanied by increased IL-6 mRNA expression in the hypothalamus. Together, these data implicate neuroinflammation as a potential contributor to sleep disruption after chemotherapy.
Collapse
Affiliation(s)
- Jeremy C Borniger
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| | - Monica M Gaudier-Diaz
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Ning Zhang
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Randy J Nelson
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - A Courtney DeVries
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
128
|
Cullen SP, Kearney CJ, Clancy DM, Martin SJ. Diverse Activators of the NLRP3 Inflammasome Promote IL-1β Secretion by Triggering Necrosis. Cell Rep 2015; 11:1535-48. [PMID: 26027935 DOI: 10.1016/j.celrep.2015.05.003] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 03/21/2015] [Accepted: 05/01/2015] [Indexed: 12/15/2022] Open
Abstract
The NLRP3 inflammasome is involved in caspase-1-dependent maturation of IL-1β in many contexts. A two-signal model has emerged for IL-1β maturation, with LPS providing "signal I" and diverse agents such as ATP, Nigericin, streptolysin O, uric acid crystals, and alum salts capable of acting as "signal II." In the absence of signal II, pro-IL-1β is upregulated but typically fails to be processed or released. What unites signal II stimuli has been debated, with the ability to promote K+ efflux suggested as a common factor, but the mechanism of IL-1β release remains unclear. Here, we show that all examined inflammasome signal II agents triggered necrosis, which was highly correlated with their ability to promote IL-1β release. IL-1β secretion occurred in tandem with the release of many additional proteins and was confined to necrotic cells. Thus, signal II agents initiate inflammation by promoting necrosis-driven IL-1β release, suggesting that IL-1β represents an inducible danger signal.
Collapse
Affiliation(s)
- Sean P Cullen
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland; Immunology Research Centre, Trinity College, Dublin 2, Ireland
| | - Conor J Kearney
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland
| | - Danielle M Clancy
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland
| | - Seamus J Martin
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland; Immunology Research Centre, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
129
|
Ran S. The Role of TLR4 in Chemotherapy-Driven Metastasis. Cancer Res 2015; 75:2405-10. [PMID: 25998620 DOI: 10.1158/0008-5472.can-14-3525] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/12/2015] [Indexed: 12/16/2022]
Abstract
Tumor resistance to cytotoxic drugs is one of the main obstacles to successful cancer therapy. Emerging evidence suggests that chemoresistance is promoted by substances released from dead and damaged cells that activate the host repair program orchestrated by Toll-like receptor-4 (TLR4). TLR4 is often overexpressed in malignant and tumor-infiltrating immune cells. In addition to endogenous ligands released by therapy-induced tumor destruction, TLR4 is directly activated by paclitaxel, one of the most commonly used chemotherapeutic drugs against various human cancers. TLR4 activation promotes local and systemic inflammation, leading to induction of multiple circuits that create a regenerative environment favoring local recurrence and metastasis. Of particular importance is TLR4-mediated recruitment of endothelial progenitors derived from immature myeloid cells. These cells play a major role in rebuilding tumor-associated lymphatic and blood vessels, thereby promoting lymphatic and hematogenous metastasis. The latter is further enhanced by the premetastatic niche generated by mobilization of myeloid provascular cells to distant organs. This review summarizes the recent evidence demonstrating that paclitaxel and other clinically used anticancer drugs actively induce metastasis even while shrinking the primary tumor. Better understanding of the mechanisms underlying TLR4-dependent chemotherapy-driven metastasis might be the key to overcoming challenges of cancer eradication.
Collapse
Affiliation(s)
- Sophia Ran
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois.
| |
Collapse
|
130
|
Toldo S, Mezzaroma E, Mauro AG, Salloum F, Van Tassell BW, Abbate A. The inflammasome in myocardial injury and cardiac remodeling. Antioxid Redox Signal 2015; 22:1146-61. [PMID: 25330141 DOI: 10.1089/ars.2014.5989] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
SIGNIFICANCE An inflammatory response follows an injury of any nature, and while such a response is an attempt to promote healing, it may, itself, result in further injury. RECENT ADVANCES The inflammasome is a macromolecular structure recently recognized as a central mediator in the acute inflammatory response. The inflammasome senses the injury and it amplifies the response by leading to the release of powerful pro-inflammatory cytokines, interleukin-1β (IL-1β) and IL-18. CRITICAL ISSUES The activation of the inflammasome in the heart during ischemic and nonischemic injury represents an exaggerated response to sterile injury and promotes adverse cardiac remodeling and failure. FUTURE DIRECTIONS Pilot clinical trials have explored blockade of the inflammasome-derived IL-1β and have shown beneficial effects on cardiac function. Additional clinical studies testing this approach are warranted. Moreover, specific inflammasome inhibitors that are ready for clinical use are currently lacking.
Collapse
Affiliation(s)
- Stefano Toldo
- 1 VCU Pauley Heart Center, Virginia Commonwealth University , Richmond, Virginia
| | | | | | | | | | | |
Collapse
|
131
|
Hassan MH, Bahashawan SA, Abdelghany TM, Abd-Allah GM, Ghobara MM. Crocin Abrogates Carbon Tetrachloride-Induced Renal Toxicity in Rats via Modulation of Metabolizing Enzymes and Diminution of Oxidative Stress, Apoptosis, and Inflammatory Cytokines. J Biochem Mol Toxicol 2015; 29:330-9. [PMID: 25899501 DOI: 10.1002/jbt.21702] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 02/07/2015] [Accepted: 03/01/2015] [Indexed: 12/22/2022]
Abstract
This work aimed at investigating the potential modulatory effects and mechanisms of crocin against CCl4 -induced nephrotoxicity. Forty male rats were allocated for three weeks treatment with corn oil, CCl4 , crocin, or crocin plus CCl4 . Crocin effectively mitigated CCl4 -induced kidney injury as evidenced by amelioration of alterations in kidney histopathology, renal weight/100 g body weight ratio and kidney functions. Crocin modulated CCl4 -induced disturbance of kidney cytochrom-P450 subfamily 2E1 and glutathione-S-transferase. The attenuation of crocin to kidney injury was also associated with suppression of oxidative stress via reduction of lipid peroxides along with induction of renal glutathione content and enhancement of superoxide dismutase, glutathione peroxidase, and catalase activities. Crocin mitigated CCl4 -induced elevation of the renal levels of tumor necrosis factor-alpha, interleukin-6, prostaglandin E2, and active caspases-3. Collectively, crocin alleviated CCl4 -induced renal damage via modulation of kidney metabolizing enzymes, suppression of oxidative stress, inhibition of inflammatory cytokines, PGE2, and active caspase3 in kidney.
Collapse
Affiliation(s)
- Memy H Hassan
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, El-Madinah El-Munaworah, Kingdom of Saudi Arabia.,Department of Pharmacology and Toxicology, Faculty of Pharmacy Al-Azahr University, Cairo, Egypt
| | - Saleh A Bahashawan
- Department of Pharmacology and Toxicology, College of Pharmacy, Taibah University, El-Madinah El-Munaworah, Kingdom of Saudi Arabia
| | - Tamer M Abdelghany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy Al-Azahr University, Cairo, Egypt
| | - Gamil M Abd-Allah
- Department of Biochemistry, Faculty of Pharmacy Al-Azahr University, Cairo, Egypt
| | - Mohamed M Ghobara
- Department of Medical Laboratories Technology, Faculty of Applied Medical Sciences, Taibah University, El-Madinah El-Munaworah, Saudi Arabia.,Department of Histology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
132
|
Toldo S, Mezzaroma E, McGeough MD, Peña CA, Marchetti C, Sonnino C, Van Tassell BW, Salloum FN, Voelkel NF, Hoffman HM, Abbate A. Independent roles of the priming and the triggering of the NLRP3 inflammasome in the heart. Cardiovasc Res 2014; 105:203-12. [PMID: 25524927 DOI: 10.1093/cvr/cvu259] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
AIMS The NLRP3 inflammasome is activated in the ischaemic heart promoting caspase-1 activation, inflammation, and cell death. Ischaemic injury establishes both a priming signal (transcription of inflammasome components) and a trigger (NLRP3 activation). Whether NLRP3 activation, without priming, induces cardiac dysfunction and/or failure is unknown. The aim of this study was to assess the independent and complementary roles of the priming and the triggering signals in the heart, in the absence of ischaemia or myocardial injury. METHODS AND RESULTS We used mice with mutant NLRP3 (constitutively active), NLRP3-A350V, under the control of tamoxifen-driven expression of the Cre recombinase (Nlrp3-A350V/CreT mice). The mice were treated for 10 days with tamoxifen before measuring the activity of caspase-1, the effector enzyme in the inflammasome. Tamoxifen treatment induced the inflammasome in the spleen but not in the heart, despite expression of the mutant NLRP3-A350V. The components of the inflammasome were significantly less expressed in the heart compared with the spleen. Subclinical low-dose lipopolysaccharide (LPS; 2 mg/kg) in Nlrp3-A350V/CreT mice induced the expression of the components of the inflammasome (priming), measured using real-time PCR and western blot, leading to the formation of an active inflammasome (caspase-1 activation) in the heart and LV systolic dysfunction while low-dose LPS was insufficient to induce LV systolic dysfunction in wild-type mice (all P < 0.01 for mutant vs. wild-type mice). CONCLUSION The signalling pathway governing the inflammasome formation in the heart requires a priming signal in order for an active NLRP3 to induce caspase-1 activation and LV dysfunction.
Collapse
Affiliation(s)
- Stefano Toldo
- VCU Pauley Heart Center, Virginia Commonwealth University, Box 980281, 1220 East Broad Street, Richmond, VA 23298, USA Victoria Johnson Research Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Eleonora Mezzaroma
- VCU Pauley Heart Center, Virginia Commonwealth University, Box 980281, 1220 East Broad Street, Richmond, VA 23298, USA Victoria Johnson Research Center, Virginia Commonwealth University, Richmond, VA, USA School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Matthew D McGeough
- Division of Allergy, Immunology and Rheumatology, University of California at San Diego, La Jolla, CA, USA
| | - Carla A Peña
- Division of Allergy, Immunology and Rheumatology, University of California at San Diego, La Jolla, CA, USA
| | - Carlo Marchetti
- VCU Pauley Heart Center, Virginia Commonwealth University, Box 980281, 1220 East Broad Street, Richmond, VA 23298, USA Victoria Johnson Research Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Chiara Sonnino
- VCU Pauley Heart Center, Virginia Commonwealth University, Box 980281, 1220 East Broad Street, Richmond, VA 23298, USA Victoria Johnson Research Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Benjamin W Van Tassell
- Victoria Johnson Research Center, Virginia Commonwealth University, Richmond, VA, USA School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Fadi N Salloum
- VCU Pauley Heart Center, Virginia Commonwealth University, Box 980281, 1220 East Broad Street, Richmond, VA 23298, USA
| | - Norbert F Voelkel
- School of Pharmacy, Virginia Commonwealth University, Richmond, VA, USA
| | - Hal M Hoffman
- Division of Allergy, Immunology and Rheumatology, University of California at San Diego, La Jolla, CA, USA
| | - Antonio Abbate
- VCU Pauley Heart Center, Virginia Commonwealth University, Box 980281, 1220 East Broad Street, Richmond, VA 23298, USA Victoria Johnson Research Center, Virginia Commonwealth University, Richmond, VA, USA
| |
Collapse
|
133
|
Braun TP, Szumowski M, Levasseur PR, Grossberg AJ, Zhu X, Agarwal A, Marks DL. Muscle atrophy in response to cytotoxic chemotherapy is dependent on intact glucocorticoid signaling in skeletal muscle. PLoS One 2014; 9:e106489. [PMID: 25254959 PMCID: PMC4177815 DOI: 10.1371/journal.pone.0106489] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 08/04/2014] [Indexed: 11/30/2022] Open
Abstract
Cancer cachexia is a syndrome of weight loss that results from the selective depletion of skeletal muscle mass and contributes significantly to cancer morbidity and mortality. The driver of skeletal muscle atrophy in cancer cachexia is systemic inflammation arising from both the cancer and cancer treatment. While the importance of tumor derived inflammation is well described, the mechanism by which cytotoxic chemotherapy contributes to cancer cachexia is relatively unexplored. We found that the administration of chemotherapy to mice produces a rapid inflammatory response. This drives activation of the hypothalamic-pituitary-adrenal axis, which increases the circulating level of corticosterone, the predominant endogenous glucocorticoid in rodents. Additionally, chemotherapy administration results in a significant loss of skeletal muscle mass 18 hours after administration with a concurrent induction of genes involved with the ubiquitin proteasome and autophagy lysosome systems. However, in mice lacking glucocorticoid receptor expression in skeletal muscle, chemotherapy-induced muscle atrophy is completely blocked. This demonstrates that cytotoxic chemotherapy elicits significant muscle atrophy driven by the production of endogenous glucocorticoids. Further, it argues that pharmacotherapy targeting the glucocorticoid receptor, given in concert with chemotherapy, is a viable therapeutic strategy in the treatment of cancer cachexia.
Collapse
Affiliation(s)
- Theodore P. Braun
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- MD/PhD Program, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Marek Szumowski
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Peter R. Levasseur
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Aaron J. Grossberg
- Department of Internal Medicine, Providence St. Vincent Medical Center, Portland, Oregon, United States of America
| | - XinXia Zhu
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Anupriya Agarwal
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, United States of America
- Division of Hematology & Medical Oncology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Daniel L. Marks
- Papé Family Pediatric Research Institute, Oregon Health & Science University, Portland, Oregon, United States of America
| |
Collapse
|
134
|
Wong J, Tran LT, Magun EA, Magun BE, Wood LJ. Production of IL-1β by bone marrow-derived macrophages in response to chemotherapeutic drugs: synergistic effects of doxorubicin and vincristine. Cancer Biol Ther 2014; 15:1395-403. [PMID: 25046000 PMCID: PMC4130732 DOI: 10.4161/cbt.29922] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cytotoxic chemotherapeutic drugs, especially when used in combination, are widely employed to treat a variety of cancers in patients but often lead to serious symptoms that negatively affect physical functioning and quality of life. There is compelling evidence that implicates cytotoxic chemotherapy-induced inflammation in the etiology of these symptoms. Because IL-1β plays a central role as an initiator cytokine in immune responses, we compared doxorubicin, a drug known to induce IL-1β production, with ten other commonly prescribed chemotherapeutic drugs in their ability to lead to processing and secretion of IL-1β by primary mouse macrophages. Seven of them (melphalan, cisplatin, vincristine, etoposide, paclitaxel, methotrexate, and cytarabine) caused the production of IL-1β in cells pretreated with lipopolysaccharide. When delivered in combination with doxorubicin, one of the drugs, vincristine, was also capable of synergistically activating the NLRP3-dependent inflammasome and increasing expression of IL-1β, IL-6, and CXCL1. The absence of TNF-α and IL-1 signaling caused a partial reduction in the production of mature IL-1β. Three small-molecule inhibitors known to suppress activity of kinases situated upstream of mitogen-activated kinases (MAPKs) inhibited the expression of IL-1β, IL-6, and CXCL1 when doxorubicin and vincristine were used singly or together, so specific kinase inhibitors may be useful in reducing inflammation in patients receiving chemotherapy.
Collapse
Affiliation(s)
- John Wong
- School of Nursing; Massachusetts General Hospital Institute of Health Professions; Boston, MA USA
| | - Lisa T Tran
- School of Nursing; Massachusetts General Hospital Institute of Health Professions; Boston, MA USA
| | - Eli A Magun
- Department of Dermatology; Oregon Health & Science University; Portland, OR USA
| | - Bruce E Magun
- School of Nursing; Massachusetts General Hospital Institute of Health Professions; Boston, MA USA
| | - Lisa J Wood
- School of Nursing; Massachusetts General Hospital Institute of Health Professions; Boston, MA USA
| |
Collapse
|
135
|
|
136
|
Vyas D, Laput G, Vyas AK. Chemotherapy-enhanced inflammation may lead to the failure of therapy and metastasis. Onco Targets Ther 2014; 7:1015-1023. [PMID: 24959088 PMCID: PMC4061164 DOI: 10.2147/ott.s60114] [Citation(s) in RCA: 229] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The lack of therapy and the failure of existing therapy has been a challenge for clinicians in treating various cancers. Doxorubicin, 5-fluorouracil, cisplatin, and paclitaxel are the first-line therapy in various cancers; however, toxicity, resistance, and treatment failure limit their clinical use. Their status leads us to discover and investigate more targeted therapy with more efficacy. In this article, we dissect literature from the patient perspective, the tumor biology perspective, therapy-induced metastasis, and cell data generated in the laboratory.
Collapse
Affiliation(s)
- Dinesh Vyas
- College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | - Gieric Laput
- College of Human Medicine, Michigan State University, East Lansing, MI, USA
| | | |
Collapse
|
137
|
Hassan MH, Ghobara M, Abd-Allah GM. Modulator Effects of Meloxicam against Doxorubicin-Induced Nephrotoxicity in Mice. J Biochem Mol Toxicol 2014; 28:337-46. [DOI: 10.1002/jbt.21570] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Revised: 04/04/2014] [Accepted: 04/07/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Memy H. Hassan
- Department of Pharmacology and Toxicology; College of Pharmacy; Taibah University; El-Madinah El-Munaworah; P.O. Box 30001 Saudi Arabia
- Department of Pharmacology and Toxicology; Faculty of Pharmacy; Al-Azahr University; Cairo Egypt
| | - Mohamed Ghobara
- Department of Medical Laboratories Technology; Faculty of Applied Medical Sciences; Taibah University; El-Madinah El-Munaworah P.O. Box 30001 Saudi Arabia
- Department of Histology; Faculty of Medicine; Tanta University; Tanta Egypt
| | - Gamil M. Abd-Allah
- Department of Biochemistry; Faculty of Pharmacy; Al-Azahr University; Cairo Egypt
| |
Collapse
|
138
|
Smith LB, Leo MC, Anderson C, Wright TJ, Weymann KB, Wood LJ. The role of IL-1β and TNF-α signaling in the genesis of cancer treatment related symptoms (CTRS): a study using cytokine receptor-deficient mice. Brain Behav Immun 2014; 38:66-76. [PMID: 24412646 PMCID: PMC3989411 DOI: 10.1016/j.bbi.2013.12.022] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/19/2013] [Accepted: 12/28/2013] [Indexed: 02/01/2023] Open
Abstract
Cytotoxic chemotherapeutic agents often induce a cluster of cancer treatment related symptoms (CTRS). The purpose of this study was to develop a mouse model of CTRS to examine the role of IL-1β and TNF-α signaling in the genesis of these symptoms. CTRS (change in wheel running activity, food intake, and body weight from baseline) were examined in wild type (WT) mice or mice lacking the TNF-α p55 (type 1) receptor (TNFR1-/-) and/or IL-1β type 1 receptor (IL-1R1-/-) injected with four doses of cyclophosphamide/Adriamycin/5-fluorouracil (CAF) at 20-day intervals. Inflammatory cytokines in blood and tissues were measured using multiplex immunoassays and quantitative RT-PCR. ANOVA was used to examine differences between genotype and/or treatment group. Kaplan-Meier analysis was used to estimate survival rate. CAF rapidly increased IL-1β and TNF-α signaling in WT mice. CAF induced acute CTRS immediately following drug injection which returned to baseline prior to the next CAF dose. Persistent CTRS were evident 3weeks after the 4th CAF dose. Acute but not persistent CTRS were associated with increased levels of IL-7, IL-9, KC, MCP-1, GCSF, and IP-10. This CAF induced inflammatory response was blunted in IL-1R1 deficient mice and absent in IL-1R1/TNFR1-deficient mice. IL-1R1-/- mice showed an identical pattern of CTRS to their WT counterparts. The assessment of CTRS in IL-1R1/TNF-R1-deficient mice was precluded by severe toxicity. Our data suggest that an important function of the IL-1β and TNF-α driven inflammatory cascade is to promote recovery following exposure to cytotoxic agents.
Collapse
Affiliation(s)
- Logan B. Smith
- School of Nursing, Oregon Health Science University, Portland, OR, 97239
| | - Michael C. Leo
- Center for Health Research, Kaiser Permanente Northwest, Portland, OR 97227
| | - Caroline Anderson
- School of Nursing, Oregon Health Science University, Portland, OR 97239, United States.
| | - Teresa J. Wright
- School of Nursing, Oregon Health Science University, Portland, OR, 97239
| | | | - Lisa J. Wood
- School of Nursing, Oregon Health Science University, Portland, OR, 97239
| |
Collapse
|
139
|
Weymann KB, Wood LJ, Zhu X, Marks DL. A role for orexin in cytotoxic chemotherapy-induced fatigue. Brain Behav Immun 2014; 37:84-94. [PMID: 24216337 PMCID: PMC3951615 DOI: 10.1016/j.bbi.2013.11.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 10/16/2013] [Accepted: 11/01/2013] [Indexed: 02/08/2023] Open
Abstract
Fatigue is the most common symptom related to cytotoxic chemotherapeutic treatment of cancer. Peripheral inflammation associated with cytotoxic chemotherapy is likely a causal factor of fatigue. The neural mechanisms by which cytotoxic chemotherapy associated inflammation induces fatigue behavior are not known. This lack of knowledge hinders development of interventions to reduce or prevent this disabling symptom. Infection induced fatigue/lethargy in rodents is mediated by suppression of hypothalamic orexin activity. Orexin is critical for maintaining wakefulness and motivated behavior. Though there are differences between infection and cytotoxic chemotherapy in some symptoms, both induce peripheral inflammation and fatigue. Based on these similarities we hypothesized that cytotoxic chemotherapy induces fatigue by disrupting orexin neuron activity. We found that a single dose of a cytotoxic chemotherapy cocktail (cyclophosphamide, adriamycin, 5-fluorouracil - CAF) induced fatigue/lethargy in mice and rats as evidenced by a significant decline in voluntary locomotor activity measured by telemetry. CAF induced inflammatory gene expression - IL-1R1 (p<0.001), IL-6 (p<0.01), TNFα (p<0.01), and MCP-1 (p<0.05) - in the rodent hypothalamus 6-24h after treatment during maximum fatigue/lethargy. CAF decreased orexin neuron activity as reflected by decreased nuclear cFos localization in orexin neurons 24h after treatment (p<0.05) and by decreased orexin-A in cerebrospinal fluid 16 h after treatment (p<0.001). Most importantly, we found that central administration of 1 μg orexin-A restored activity in CAF-treated rats (p<0.05). These results demonstrate that cytotoxic chemotherapy induces hypothalamic inflammation and that suppression of hypothalamic orexin neuron activity has a causal role in cytotoxic chemotherapy-induced fatigue in rodents.
Collapse
Affiliation(s)
- K B Weymann
- Oregon Health & Science University, Portland, OR 97239, United States.
| | - L J Wood
- Oregon Health & Science University, Portland, OR 97239, United States; School of Nursing, MGH Institute of Health Professions, Boston, MA 02129, United States.
| | - X Zhu
- Oregon Health & Science University, Portland, OR 97239, United States.
| | - D L Marks
- Oregon Health & Science University, Portland, OR 97239, United States.
| |
Collapse
|
140
|
Animal models in studies of cardiotoxicity side effects from antiblastic drugs in patients and occupational exposed workers. BIOMED RESEARCH INTERNATIONAL 2014; 2014:240642. [PMID: 24701565 PMCID: PMC3950409 DOI: 10.1155/2014/240642] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 10/29/2013] [Accepted: 11/07/2013] [Indexed: 01/08/2023]
Abstract
Cardiotoxicity is an important side effect of cytotoxic drugs and may be a risk factor of long-term morbidity for both patients during therapy and also for staff exposed during the phases of manipulation of antiblastic drugs. The mechanism of cardiotoxicity studied in vitro and in vivo essentially concerns the formation of free radicals leading to oxidative stress, with apoptosis of cardiac cells or immunologic reactions, but other mechanisms may play a role in antiblastic-induced cardiotoxicity. Actually, some new cytotoxic drugs like trastuzumab and cyclopentenyl cytosine show cardiotoxic effects. In this report we discuss the different mechanisms of cardiotoxicity induced by antiblastic drugs assessed using animal models.
Collapse
|
141
|
Antonopoulos C, Dubyak GR. Chemotherapy engages multiple pathways leading to IL-1β production by myeloid leukocytes. Oncoimmunology 2014; 3:e27499. [PMID: 24800164 PMCID: PMC4006851 DOI: 10.4161/onci.27499] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 12/12/2013] [Indexed: 12/13/2022] Open
Abstract
Interleukin-1β (IL-1β) can limit tumor growth by promoting T cell-mediated antitumor immune responses. Several chemotherapeutic agents can stimulate the production of IL-1β by tumor-infiltrating leukocytes via the NLRP3 inflammasome. We have recently demonstrated that some chemotherapeutics can also trigger the secretion of IL-1β by driving the assembly of the caspase-8- and FADD-containing platform known as the ripoptosome.
Collapse
Affiliation(s)
- Christina Antonopoulos
- Department of Pathology; Case Western Reserve University School of Medicine; Cleveland, OH USA
| | - George R Dubyak
- Department of Physiology and Biophysics; Case Western Reserve University School of Medicine; Cleveland, OH USA ; Department of Pathology; Case Western Reserve University School of Medicine; Cleveland, OH USA ; Case Comprehensive Cancer Center; Case Western Reserve University School of Medicine; Cleveland, OH USA
| |
Collapse
|
142
|
Figueiredo N, Chora A, Raquel H, Pejanovic N, Pereira P, Hartleben B, Neves-Costa A, Moita C, Pedroso D, Pinto A, Marques S, Faridi H, Costa P, Gozzelino R, Zhao JL, Soares MP, Gama-Carvalho M, Martinez J, Zhang Q, Döring G, Grompe M, Simas JP, Huber TB, Baltimore D, Gupta V, Green DR, Ferreira JA, Moita LF. Anthracyclines induce DNA damage response-mediated protection against severe sepsis. Immunity 2013; 39:874-84. [PMID: 24184056 DOI: 10.1016/j.immuni.2013.08.039] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Accepted: 08/13/2013] [Indexed: 12/24/2022]
Abstract
Severe sepsis remains a poorly understood systemic inflammatory condition with high mortality rates and limited therapeutic options in addition to organ support measures. Here we show that the clinically approved group of anthracyclines acts therapeutically at a low dose regimen to confer robust protection against severe sepsis in mice. This salutary effect is strictly dependent on the activation of DNA damage response and autophagy pathways in the lung, as demonstrated by deletion of the ataxia telangiectasia mutated (Atm) or the autophagy-related protein 7 (Atg7) specifically in this organ. The protective effect of anthracyclines occurs irrespectively of pathogen burden, conferring disease tolerance to severe sepsis. These findings demonstrate that DNA damage responses, including the ATM and Fanconi Anemia pathways, are important modulators of immune responses and might be exploited to confer protection to inflammation-driven conditions, including severe sepsis.
Collapse
Affiliation(s)
- Nuno Figueiredo
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; Clínica Universitária de Cirurgia I, Centro Hospitalar Lisboa Norte, EPE, 1649-028 Lisboa, Portugal; Gulbenkian Programme for Advanced Medical Education, 2780-156 Oeiras, Portugal; Champalimaud Foundation, 1400-038 Lisboa, Portugal
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Antonopoulos C, El Sanadi C, Kaiser WJ, Mocarski ES, Dubyak GR. Proapoptotic chemotherapeutic drugs induce noncanonical processing and release of IL-1β via caspase-8 in dendritic cells. THE JOURNAL OF IMMUNOLOGY 2013; 191:4789-803. [PMID: 24078693 DOI: 10.4049/jimmunol.1300645] [Citation(s) in RCA: 188] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The identification of noncanonical (caspase-1-independent) pathways for IL-1β production has unveiled an intricate interplay between inflammatory and death-inducing signaling platforms. We found a heretofore unappreciated role for caspase-8 as a major pathway for IL-1β processing and release in murine bone marrow-derived dendritic cells (BMDC) costimulated with TLR4 agonists and proapoptotic chemotherapeutic agents such as doxorubicin (Dox) or staurosporine (STS). The ability of Dox to stimulate release of mature (17-kDa) IL-1β was nearly equivalent in wild-type (WT) BMDC, Casp1(-/-)Casp11(-/-) BMDC, WT BMDC treated with the caspase-1 inhibitor YVAD, and BMDC lacking the inflammasome regulators ASC, NLRP3, or NLRC4. Notably, Dox-induced production of mature IL-1β was temporally correlated with caspase-8 activation in WT cells and greatly suppressed in Casp8(-/-)Rip3(-/-) or Trif(-/-) BMDC, as well as in WT BMDC treated with the caspase-8 inhibitor, IETD. Similarly, STS stimulated robust IL-1β processing and release in Casp1(-/-)Casp11(-/-) BMDC that was IETD sensitive. These data suggest that TLR4 induces assembly of caspase-8-based signaling complexes that become licensed as IL-1β-converting enzymes in response to Dox and STS. The responses were temporally correlated with downregulation of cellular inhibitor of apoptosis protein 1, suggesting suppressive roles for this and likely other inhibitor of apoptosis proteins on the stability and/or proteolytic activity of the caspase-8 platforms. Thus, proapoptotic chemotherapeutic agents stimulate the caspase-8-mediated processing and release of IL-1β, implicating direct effects of such drugs on a noncanonical inflammatory cascade that may modulate immune responses in tumor microenvironments.
Collapse
Affiliation(s)
- Christina Antonopoulos
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | | | | | | | | |
Collapse
|
144
|
L'homme L, Esser N, Riva L, Scheen A, Paquot N, Piette J, Legrand-Poels S. Unsaturated fatty acids prevent activation of NLRP3 inflammasome in human monocytes/macrophages. J Lipid Res 2013; 54:2998-3008. [PMID: 24006511 DOI: 10.1194/jlr.m037861] [Citation(s) in RCA: 121] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The NLRP3 inflammasome is involved in many obesity-associated diseases, such as type 2 diabetes, atherosclerosis, and gouty arthritis, through its ability to induce interleukin (IL)-1β release. The molecular link between obesity and inflammasome activation is still unclear, but free fatty acids have been proposed as one triggering event. Here we reported opposite effects of saturated fatty acids (SFAs) compared with unsaturated fatty acids (UFAs) on NLRP3 inflammasome in human monocytes/macrophages. Palmitate and stearate, both SFAs, triggered IL-1β secretion in a caspase-1/ASC/NLRP3-dependent pathway. Unlike SFAs, the UFAs oleate and linoleate did not lead to IL-1β secretion. In addition, they totally prevented the IL-1β release induced by SFAs and, with less efficiency, by a broad range of NLRP3 inducers, including nigericin, alum, and monosodium urate. UFAs did not affect the transcriptional effect of SFAs, suggesting a specific effect on the NLRP3 activation. These results provide a new anti-inflammatory mechanism of UFAs by preventing the activation of the NLRP3 inflammasome and, therefore, IL-1β processing. By this way, UFAs might play a protective role in NLRP3-associated diseases.
Collapse
Affiliation(s)
- Laurent L'homme
- Laboratory of Virology and Immunology, GIGA-Signal Transduction
| | | | | | | | | | | | | |
Collapse
|
145
|
Pal S, Shankar BS, Sainis KB. Cytokines from the tumor microenvironment modulate sirtinol cytotoxicity in A549 lung carcinoma cells. Cytokine 2013; 64:196-207. [PMID: 23972545 DOI: 10.1016/j.cyto.2013.07.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 07/15/2013] [Accepted: 07/30/2013] [Indexed: 11/19/2022]
Abstract
Cytokines in tumor microenvironment play an important role in the success or failure of molecular targeted therapies. We have chosen tumor necrosis factor α (TNF-α), TNF related apoptosis inducing ligand (TRAIL), insulin-like growth factor 1 (IGF-1) and transforming growth factor β (TGF-β) as representative pro-inflammatory, pro-apoptotic, anti-apoptotic and anti-inflammatory tumor derived cytokines. Analysis of Oncomine database revealed the differential expression of these cytokines in a subset of cancer patients. The effects of these cytokines on cytotoxicity of FDA approved drugs - cisplatin and taxol and inhibitors of epidermal growth factor receptor - AG658, Janus kinase - AG490 and SIRT1 - sirtinol were assessed in A549 lung cancer cells. TRAIL augmented cytotoxicity of sirtinol and IGF-1 had a sparing effect. Since TRAIL and IGF-1 differentially modulated sirtinol cytotoxicity, further studies were carried out to identify the mechanisms. Sirtinol or knockdown of SIRT1 increased the expression of death receptors DR4 and DR5 and sensitized A549 cells to TRAIL. Increased cell death in presence of TRAIL and sirtinol was caspase independent and demonstrated classical features of necroptosis. Inhibition of iNOS increased caspase activity and switched the mode of cell death to caspase mediated apoptosis. Interestingly, sirtinol or SIRT1 knockdown did not increase IGF-1R expression. Instead, it abrogated ligand induced downregulation of IGF-1R and increased cell survival through PI3K-AKT pathway. In conclusion, these findings reveal that the tumor microenvironment contributes to modulation of cytotoxicity of drugs and that combination therapy, with agents that increase TRAIL signaling and suppress IGF-1 pathway may potentiate anticancer effect.
Collapse
Affiliation(s)
- Shyama Pal
- Radiation Biology & Health Sciences Division, Bio-Medical Group, Bhabha Atomic Research Centre, Modular Laboratories, Mumbai 400085, India
| | | | | |
Collapse
|
146
|
Abstract
Cardiac toxicity is a major dose-limiting factor for the anthracycline drug doxorubicin. The reasons why doxorubicin causes heart damage are not fully understood, and the manuscript by Wong et al. postulates that inflammatory cytokines released from macrophages or other cell types may play a significant role in the damage process in response to doxorubicin and possibly other chemotherapeutic agents.1 Expression of many cytokines requires activation of both the p38 MAPK and JNK pathways and, additionally, doxorubicin toxicity can be blocked by combined inhibition of both pathways.2,3 The MAP3K responsible for doxorubicin-induced p38 MAPK and JNK activation in keratinocytes was previously shown by these authors to be ZAK.4 ZAK is of note because it can be targeted by FDA approved agents such as nilotinib and sorafenib.4-7
Collapse
Affiliation(s)
- Paul Dent
- Department of Neurosurgery; Massey Cancer Center; Virginia Commonwealth University; Richmond, VA USA
| |
Collapse
|
147
|
Inflammation and neural signaling: etiologic mechanisms of the cancer treatment-related symptom cluster. Curr Opin Support Palliat Care 2013; 7:54-9. [PMID: 23314015 DOI: 10.1097/spc.0b013e32835dabe3] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Cancer patients undergoing treatment with cytotoxic chemotherapeutic agents (CCAs) often experience a cluster of treatment-related symptoms, which include fatigue, loss of appetite, disturbed sleep, depressed mood, cognitive difficulties, and changes in body composition. This symptom cluster collectively referred to herein as cancer treatment-related symptoms (CTRSs) decrease quality of life, and physical and social functioning. The preclinical and clinical studies described in this review represent important progress in understanding potential underlying mechanisms of CTRS. RECENT FINDINGS Recent studies support a role for CCA-induced interleukin-1β (IL-1β) signaling in the cause of CTRS. CCAs may share a common ability to activate intracellular stress response pathways to trigger the synthesis, processing, and release of IL-1β from immune cells. Fatigue, sleep disturbance, and cognitive difficulties in cancer patients exposed to CCAs correlate with plasma levels of IL-6, IL-1 receptor antagonist, and soluble tumor necrosis factor receptor-I/II, surrogate markers of IL-1β-mediated central nervous system (CNS) inflammation. Additional preclinical work suggests IL-1β-mediated CNS inflammation may cause CTRS by altering hypothalamic and hippocampal functioning. SUMMARY Although additional research is necessary to further establish the link between CCA exposure, IL-1β-mediated inflammatory processes and CTRS, these data provide hints for future studies and therapeutic approaches in ameliorating these symptoms in cancer patients.
Collapse
|
148
|
Wang SZ, He H, Han R, Zhu JL, Kou JQ, Ding XL, Qin ZH. The Protective Effects of Cobra Venom from Naja naja atra on Acute and Chronic Nephropathy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2013; 2013:478049. [PMID: 23983784 PMCID: PMC3745861 DOI: 10.1155/2013/478049] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 05/23/2013] [Accepted: 05/23/2013] [Indexed: 12/25/2022]
Abstract
This study investigated the effects of Naja naja atra venom (NNAV) on acute and chronic nephropathy in rats. Rats received 6 mg/kg adriamycin (ADR) once to evoke the chronic nephropathy or 8 ml/kg 50% v/v glycerol to produce acute renal failure (ARF). The NNAV was given orally once a day starting five days prior to ADR or glycerol injection and continued to the end of experiments. The animals were placed in metabolic cages for 24 h for urine collection for urinary protein determination. The kidney function-related biochemical changes and index of oxidative stress were determined with automatic biochemistry analyzer or colorimetric enzyme assay kits. The pathomorphological changes were observed using light and transmission electron microcopies. The levels of inflammatory cytokines and NF- κ B activation were determined using ELISA kits, Western blot analysis, or immunofluorescence. The results showed that NNAV relieved ADR-induced chronic nephropathy and glycerol-triggered acute renal failure syndromes including proteinuria, hypoalbuminemia, hyperlipidemia, serum electrolyte unbalance, renal oxidative stress, and pathological damages. NNAV reduced kidney levels of TNF- α and IL-1 β , but it increased the levels of I κ B- α and inhibited NF- κ B p65 nuclear localization. These findings suggest that NNAV may be a valuable therapeutic drug for acute and chronic kidney diseases.
Collapse
Affiliation(s)
- Shu-Zhi Wang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou 215123, China
| | - He He
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou 215123, China
| | - Rong Han
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou 215123, China
| | - Jia-Li Zhu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou 215123, China
| | - Jian-Qun Kou
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou 215123, China
| | - Xiao-Lan Ding
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou 215123, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, 199 Ren Ai Road, Suzhou 215123, China
| |
Collapse
|
149
|
Guo RM, Xu WM, Lin JC, Mo LQ, Hua XX, Chen PX, Wu K, Zheng DD, Feng JQ. Activation of the p38 MAPK/NF-κB pathway contributes to doxorubicin-induced inflammation and cytotoxicity in H9c2 cardiac cells. Mol Med Rep 2013; 8:603-8. [PMID: 23807148 DOI: 10.3892/mmr.2013.1554] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 06/18/2013] [Indexed: 11/06/2022] Open
Abstract
A number of studies have demonstrated that inflammation plays a role in doxorubicin (DOX)-induced cardiotoxicity. However, the molecular mechanism by which DOX induces cardiac inflammation has yet to be fully elucidated. The present study aimed to investigate the role of the p38 mitogen-activated protein kinase (MAPK)/nuclear factor-κB (NF-κB) pathway in DOX-induced inflammation and cytotoxicity. The results of our study demonstrated that the exposure of H9c2 cardiac cells to DOX reduced cell viability and stimulated an inflammatory response, as demonstrated by an increase in the levels of interleukin-1β (IL-1β) and IL-6, as well as tumor necrosis factor-α (TNF-α) production. Notably, DOX exposure induced the overexpression of phosphorylated p38 MAPK and phosphorylation of the NF-κB p65 subunit, which was markedly inhibited by SB203580, a specific inhibitor of p38 MAPK. The inhibition of NF-κB by pyrrolidine dithiocarbamate (PDTC), a selective inhibitor of NF-κB, significantly ameliorated DOX-induced inflammation, leading to a decrease in the levels of IL-1β and IL-6, as well as TNF-α production in H9c2 cells. The pretreatment of H9c2 cells with either SB203580 or PDTC before exposure to DOX significantly attenuated DOX-induced cytotoxicity. In conclusion, our study provides novel data demonstrating that the p38 MAPK/NF-κB pathway is important in the induction of DOX-induced inflammation and cytotoxicity in H9c2 cardiac myocytes.
Collapse
Affiliation(s)
- Run-Min Guo
- Department of Physiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
150
|
Prenylated Flavonoids from Morus alba L. Cause Inhibition of G1/S Transition in THP-1 Human Leukemia Cells and Prevent the Lipopolysaccharide-Induced Inflammatory Response. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:350519. [PMID: 23762124 PMCID: PMC3671669 DOI: 10.1155/2013/350519] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 04/26/2013] [Indexed: 12/04/2022]
Abstract
Morus alba L. (MA) is a natural source of many compounds with different biological effects. It has been described to possess anti-inflammatory, antioxidant, and hepatoprotective activities. The aim of this study was to evaluate cytotoxicity of three flavonoids isolated from MA (kuwanon E, cudraflavone B, and 4′-O-methylkuwanon E) and to determine their effects on proliferation of THP-1 cells, and on cell cycle progression of cancer cells. Anti-inflammatory effects were also determined for all three given flavonoids.
Methods used in the study included quantification of cells by hemocytometer and WST-1 assays, flow cytometry, western blotting, ELISA, and zymography.
From the three compounds tested, cudraflavone B showed the strongest effects on cell cycle progression and viability of tumor and/or immortalized cells and also on inflammatory response of macrophage-like cells. Kuwanon E and 4′-O-methylkuwanon E exerted more sophisticated rather than direct toxic effect on used cell types. Our data indicate that mechanisms different from stress-related or apoptotic signaling pathways are involved in the action of these compounds. Although further studies are required to precisely define the mechanisms of MA flavonoid action in human cancer and macrophage-like cells, here we demonstrate their effects combining antiproliferative and anti-inflammatory activities, respectively.
Collapse
|