1
|
Panchal J, Jaiswal S, Jain S, Kumawat J, Sharma A, Jain P, Jain S, Verma K, Dwivedi J, Sharma S. Development of novel bosentan analogues as endothelin receptor antagonists for pulmonary arterial hypertension. Eur J Med Chem 2023; 259:115681. [PMID: 37515921 DOI: 10.1016/j.ejmech.2023.115681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 07/22/2023] [Accepted: 07/23/2023] [Indexed: 07/31/2023]
Abstract
Since decades, bosentan has been in use for the treatment of pulmonary arterial hypertension (PAH). However, chronic exposure to bosentan leads to the development of resistance, tolerance, and serious adverse effects that have restricted its usage in clinical practices. To surmount these limitations, some new bosentan derivatives have been synthesized and evaluated for their therapeutic efficacy in PAH. Molecular docking analyses of all the synthesized derivatives were carried out using the endothelin (ET) receptor. In addition, the inhibitory ability of synthesized derivatives was determined in in vitro assay employing an ET-1 human ELISA kit. Among the synthesized derivatives, three derivatives namely 17d, 16j, and 16h with higher docking scores and lower IC50 values were selected for determination of the magnitude of the binding force between the derivative and ET receptor using molecular dynamics (MD) simulations study. Further, these derivatives were subjected to in vivo studies using monocrotaline (MCT) induced PAH in rat model. Results of in vivo studies inferred that the derivatives exhibit impressive ability to reduce PAH. Besides, its protective role was also evidenced in hemodynamic and right ventricular hypertrophy analyses, histological analysis, cardiac biomarkers, hypoxia-inducible factor 1 alpha (HIF1α) levels, and biochemical studies. Furthermore, gene quantification by quantitative RT-PCR and Western blot analysis was also performed to examine its effect on the expression of key proteins in PAH. Notably, amongst three, derivative 16h exhibited the most encouraging results in molecular docking analysis, in vitro, in vivo, histopathological, biochemical, protein expression, and MD studies. Besides, derivative 16h also showed impressive pharmacokinetic features in ADMET analysis. In conclusion, derivative 16 h could act as a reliable ET receptor antagonist and requires further exploration to attain its therapeutic utility in PAH management.
Collapse
Affiliation(s)
- Jigar Panchal
- Department of Chemistry, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Shivangi Jaiswal
- Department of Chemistry, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Sonika Jain
- Department of Chemistry, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India.
| | - Jyoti Kumawat
- Department of Chemistry, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Ashima Sharma
- Department of Pharmacy, Panjab University, Chandigarh, 160014, Punjab, India
| | - Pankaj Jain
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Smita Jain
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Kanika Verma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India.
| | - Swapnil Sharma
- Department of Pharmacy, Banasthali Vidyapith, Banasthali, 304022, Rajasthan, India.
| |
Collapse
|
2
|
Maru S, Verma J, Wilen CE, Rosenholm JM, Bansal KK. Attenuation of celecoxib cardiac toxicity using Poly(δ-decalactone) based nanoemulsion via oral route. Eur J Pharm Sci 2023; 190:106585. [PMID: 37717666 DOI: 10.1016/j.ejps.2023.106585] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/19/2023]
Abstract
Celecoxib (CLX), a poorly soluble anti-inflammatory drug, requires administration in higher concentrations to produce therapeutic effects, oftentimes resulting in cardiac toxicity. Therefore, in this study, we employed a nanoemulsion technology to improve the solubility of CLX using poly(δ-decalactone) (PDL) polymer as an oil and mPEG-b-PDL as a surfactant. The nanoemulsion (NE) was successfully prepared via the nanoprecipitation method. In vitro characterization was performed for size, drug release, and stability. In vivo studies were performed to establish anti-inflammatory activity, CLX induced cardiac toxicity, and pharmacokinetic profile of NE, post-oral administration. The globular size of less than 100 nm was obtained in NE with high CLX loading. The in vitro drug release studies suggested ∼90% of CLX release from NE within 96 h. A significant anti-inflammatory activity with lowered cardiac marker values was observed for CLX NE compared to a marketed drug formulation. The pharmacokinetic study revealed that the mean retention time of CLX was significantly increased with NE in contrast to the marketed formulation, suggesting the advantage of administering CLX in the form of NE owing to the higher solubility and sustained release pattern. The long-term storage stability study reveals that NE does not show significant changes in terms of size with only a slight decrement in CLX content was observed after 24 months. The obtained results indicate that CLX bioavailability has been considerably improved without being toxic to the heart with the aid of NE and advocate the use of PDL NE for developing oral formulations for poorly soluble drugs.
Collapse
Affiliation(s)
- Saurabh Maru
- School of Pharmacy and Technology Management, SVKM's NMIMS, Babulde, Banks of Tapi River, Mumbai-Agra Road, Shirpur, Maharashtra 425405, India
| | - Jyoti Verma
- Pharmaceutical Sciences Laboratory Faculty of Science and Engineering Åbo Akademi University 20520 Turku, Finland.
| | - Carl-Eric Wilen
- Laboratory of Molecular Science and Engineering, Åbo Akademi University, Aurum, Henrikinkatu 2, 20500 Turku, Finland.
| | - Jessica M Rosenholm
- Pharmaceutical Sciences Laboratory Faculty of Science and Engineering Åbo Akademi University 20520 Turku, Finland.
| | - Kuldeep K Bansal
- Pharmaceutical Sciences Laboratory Faculty of Science and Engineering Åbo Akademi University 20520 Turku, Finland; Laboratory of Molecular Science and Engineering, Åbo Akademi University, Aurum, Henrikinkatu 2, 20500 Turku, Finland.
| |
Collapse
|
3
|
Hydropersulfides (RSSH) Outperform Post-Conditioning and Other Reactive Sulfur Species in Limiting Ischemia-Reperfusion Injury in the Isolated Mouse Heart. Antioxidants (Basel) 2022; 11:antiox11051010. [PMID: 35624878 PMCID: PMC9137952 DOI: 10.3390/antiox11051010] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 01/21/2023] Open
Abstract
Hydrogen sulfide (H2S) exhibits protective effects in cardiovascular disease such as myocardial ischemia/reperfusion (I/R) injury, cardiac hypertrophy, and atherosclerosis. Despite these findings, its mechanism of action remains elusive. Recent studies suggest that H2S can modulate protein activity through redox-based post-translational modifications of protein cysteine residues forming hydropersulfides (RSSH). Furthermore, emerging evidence indicates that reactive sulfur species, including RSSH and polysulfides, exhibit cardioprotective action. However, it is not clear yet whether there are any pharmacological differences in the use of H2S vs. RSSH and/or polysulfides. This study aims to examine the differing cardioprotective effects of distinct reactive sulfur species (RSS) such as H2S, RSSH, and dialkyl trisulfides (RSSSR) compared with canonical ischemic post-conditioning in the context of a Langendorff ex-vivo myocardial I/R injury model. For the first time, a side-by-side study has revealed that exogenous RSSH donation is a superior approach to maintain post-ischemic function and limit infarct size when compared with other RSS and mechanical post-conditioning. Our results also suggest that RSSH preserves mitochondrial respiration in H9c2 cardiomyocytes exposed to hypoxia-reoxygenation via inhibition of oxidative phosphorylation while preserving cell viability.
Collapse
|
4
|
Wakefield ZR, Tanaka M, Pampo C, Lepler S, Rice L, Guingab-Cagmat J, Garrett TJ, Siemann DW. Normal tissue and tumor microenvironment adaptations to aerobic exercise enhance doxorubicin anti-tumor efficacy and ameliorate its cardiotoxicity in retired breeder mice. Oncotarget 2021; 12:1737-1748. [PMID: 34504647 PMCID: PMC8416558 DOI: 10.18632/oncotarget.28057] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/13/2021] [Indexed: 11/25/2022] Open
Abstract
Aerobic exercise is receiving increased recognition in oncology for its multiple purported benefits. Exercise is known to induce physiologic adaptations that improve patient quality-of-life parameters as well as all-cause mortality. There also is a growing body of evidence that exercise may directly alter the tumor microenvironment to influence tumor growth, metastasis, and response to anticancer therapies. Furthermore, the physiologic adaptations to exercise in normal tissues may protect against treatment-associated toxicity and allow for greater treatment tolerance. However, the exercise prescription required to induce these beneficial tumor-related outcomes remains unclear. This study characterized the aerobic adaptations to voluntary wheel running in normal tissues and the tumor microenvironment. Female, retired breeder BALB/c mice and syngeneic breast adenocarcinoma cells were utilized in primary tumor and metastasis models. Aerobic exercise was found to induce numerous adaptations across various tissues in these mice, although primary tumor growth and metastasis were largely unaffected. However, intratumoral hypoxia and global metabolism were altered in the tumors of exercising hosts relative to non-wheel running controls. Doxorubicin chemotherapy also was found to be more efficacious at delaying tumor growth with adjuvant aerobic exercise. Additionally, doxorubicin-induced cardiac toxicity was ameliorated in exercising hosts relative to non-wheel running controls. Taken together, these data suggest that the normal tissue and tumor microenvironment adaptations to aerobic exercise can improve doxorubicin efficacy while simultaneously limiting its toxicity.
Collapse
Affiliation(s)
- Zachary R Wakefield
- Department of Radiation Oncology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mai Tanaka
- Department of Radiation Oncology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Christine Pampo
- Department of Radiation Oncology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Sharon Lepler
- Department of Radiation Oncology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Lori Rice
- Department of Radiation Oncology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Joy Guingab-Cagmat
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Timothy J Garrett
- Department of Pathology, Immunology, and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Dietmar W Siemann
- Department of Radiation Oncology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
5
|
Azimzadeh O, von Toerne C, Subramanian V, Sievert W, Multhoff G, Atkinson MJ, Tapio S. Data-Independent Acquisition Proteomics Reveals Long-Term Biomarkers in the Serum of C57BL/6J Mice Following Local High-Dose Heart Irradiation. Front Public Health 2021; 9:678856. [PMID: 34277544 PMCID: PMC8283568 DOI: 10.3389/fpubh.2021.678856] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/18/2021] [Indexed: 12/23/2022] Open
Abstract
Background and Purpose: Cardiotoxicity is a well-known adverse effect of radiation therapy. Measurable abnormalities in the heart function indicate advanced and often irreversible heart damage. Therefore, early detection of cardiac toxicity is necessary to delay and alleviate the development of the disease. The present study investigated long-term serum proteome alterations following local heart irradiation using a mouse model with the aim to detect biomarkers of radiation-induced cardiac toxicity. Materials and Methods: Serum samples from C57BL/6J mice were collected 20 weeks after local heart irradiation with 8 or 16 Gy X-ray; the controls were sham-irradiated. The samples were analyzed by quantitative proteomics based on data-independent acquisition mass spectrometry. The proteomics data were further investigated using bioinformatics and ELISA. Results: The analysis showed radiation-induced changes in the level of several serum proteins involved in the acute phase response, inflammation, and cholesterol metabolism. We found significantly enhanced expression of proinflammatory cytokines (TNF-α, TGF-β, IL-1, and IL-6) in the serum of the irradiated mice. The level of free fatty acids, total cholesterol, low-density lipoprotein (LDL), and oxidized LDL was increased, whereas that of high-density lipoprotein was decreased by irradiation. Conclusions: This study provides information on systemic effects of heart irradiation. It elucidates a radiation fingerprint in the serum that may be used to elucidate adverse cardiac effects after radiation therapy.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Institute of Radiation Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Section Radiation Biology, Federal Office for Radiation Protection, Oberschleissheim, Germany
| | - Christine von Toerne
- Research Unit Protein Science, Helmholtz Zentrum München - German Research Center for Environmental Health, Munich, Germany
| | - Vikram Subramanian
- Institute of Radiation Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Wolfgang Sievert
- Department of Radiation Oncology, Center for Translational Cancer Research (TranslaTUM), Campus Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Center for Translational Cancer Research (TranslaTUM), Campus Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Michael J Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Radiation Biology, Technical University of Munich, Munich, Germany
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Institute for Biological and Medical Imaging, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
6
|
Azevedo Voltarelli V, Coronado M, Gonçalves Fernandes L, Cruz Campos J, Jannig PR, Batista Ferreira JC, Fajardo G, Chakur Brum P, Bernstein D. β 2-Adrenergic Signaling Modulates Mitochondrial Function and Morphology in Skeletal Muscle in Response to Aerobic Exercise. Cells 2021; 10:cells10010146. [PMID: 33450889 PMCID: PMC7828343 DOI: 10.3390/cells10010146] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/28/2020] [Accepted: 01/11/2021] [Indexed: 02/06/2023] Open
Abstract
The molecular mechanisms underlying skeletal muscle mitochondrial adaptations induced by aerobic exercise (AE) are not fully understood. We have previously shown that AE induces mitochondrial adaptations in cardiac muscle, mediated by sympathetic stimulation. Since direct sympathetic innervation of neuromuscular junctions influences skeletal muscle homeostasis, we tested the hypothesis that β2-adrenergic receptor (β2-AR)-mediated sympathetic activation induces mitochondrial adaptations to AE in skeletal muscle. Male FVB mice were subjected to a single bout of AE on a treadmill (80% Vmax, 60 min) under β2-AR blockade with ICI 118,551 (ICI) or vehicle, and parameters of mitochondrial function and morphology/dynamics were evaluated. An acute bout of AE significantly increased maximal mitochondrial respiration in tibialis anterior (TA) isolated fiber bundles, which was prevented by β2-AR blockade. This increased mitochondrial function after AE was accompanied by a change in mitochondrial morphology towards fusion, associated with increased Mfn1 protein expression and activity. β2-AR blockade fully prevented the increase in Mfn1 activity and reduced mitochondrial elongation. To determine the mechanisms involved in mitochondrial modulation by β2-AR activation in skeletal muscle during AE, we used C2C12 myotubes, treated with the non-selective β-AR agonist isoproterenol (ISO) in the presence of the specific β2-AR antagonist ICI or during protein kinase A (PKA) and Gαi protein blockade. Our in vitro data show that β-AR activation significantly increases mitochondrial respiration in myotubes, and this response was dependent on β2-AR activation through a Gαs-PKA signaling cascade. In conclusion, we provide evidence for AE-induced β2-AR activation as a major mechanism leading to alterations in mitochondria function and morphology/dynamics. β2-AR signaling is thus a key-signaling pathway that contributes to skeletal muscle plasticity in response to exercise.
Collapse
Affiliation(s)
- Vanessa Azevedo Voltarelli
- Department of Biodynamics of the Human Body Movement, School of Physical Education and Sport, University of São Paulo, São Paulo 05508-030, SP, Brazil; (V.A.V.); (L.G.F.); (P.R.J.)
| | - Michael Coronado
- Department of Pediatrics, School of Medicine, Stanford University, Palo Alto, CA 94304, USA; (M.C.); (G.F.)
| | - Larissa Gonçalves Fernandes
- Department of Biodynamics of the Human Body Movement, School of Physical Education and Sport, University of São Paulo, São Paulo 05508-030, SP, Brazil; (V.A.V.); (L.G.F.); (P.R.J.)
| | - Juliane Cruz Campos
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-030, SP, Brazil; (J.C.C.); (J.C.B.F.)
| | - Paulo Roberto Jannig
- Department of Biodynamics of the Human Body Movement, School of Physical Education and Sport, University of São Paulo, São Paulo 05508-030, SP, Brazil; (V.A.V.); (L.G.F.); (P.R.J.)
| | - Julio Cesar Batista Ferreira
- Department of Anatomy, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-030, SP, Brazil; (J.C.C.); (J.C.B.F.)
- Department of Chemical and Systems Biology, School of Medicine, Stanford University, Palo Alto, CA 94304, USA
| | - Giovanni Fajardo
- Department of Pediatrics, School of Medicine, Stanford University, Palo Alto, CA 94304, USA; (M.C.); (G.F.)
| | - Patricia Chakur Brum
- Department of Biodynamics of the Human Body Movement, School of Physical Education and Sport, University of São Paulo, São Paulo 05508-030, SP, Brazil; (V.A.V.); (L.G.F.); (P.R.J.)
- Correspondence: or (P.C.B.); (D.B.); Tel.: +55-11-30913136 (P.C.B.); Fax: +55-11-38135921 (P.C.B.)
| | - Daniel Bernstein
- Department of Pediatrics, School of Medicine, Stanford University, Palo Alto, CA 94304, USA; (M.C.); (G.F.)
- Correspondence: or (P.C.B.); (D.B.); Tel.: +55-11-30913136 (P.C.B.); Fax: +55-11-38135921 (P.C.B.)
| |
Collapse
|
7
|
Pentoxifylline Attenuates Arsenic Trioxide-Induced Cardiac Oxidative Damage in Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6406318. [PMID: 33505582 PMCID: PMC7810555 DOI: 10.1155/2021/6406318] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 12/09/2020] [Accepted: 12/22/2020] [Indexed: 12/30/2022]
Abstract
This study was undertaken to evaluate the therapeutic potential effect of pentoxifylline (PTX) against arsenic trioxide (ATO)-induced cardiac oxidative damage in mice. Thirty-six male albino mice were divided into six groups and treated intraperitoneally with normal saline (group 1), ATO (5 mg/kg; group 2), PTX (100 mg/kg; group 3), and different doses of PTX (25, 50, and 100 mg/kg; groups 4, 5, and 6, respectively) with ATO. After four weeks, the blood sample was collected for biochemical experiments. In addition, cardiac tissue was removed for assessment of oxidative stress markers and histopathological changes (such as hemorrhage, necrosis, infiltration of inflammatory cells, and myocardial degeneration). The findings showed that ATO caused a significant raise in serum biochemical markers such as lactate dehydrogenase (LDH), creatine phosphokinase (CPK) and troponin-I (cTnI), glucose, total cholesterol (TC), and triglyceride (TG) levels. In addition to histopathological changes in cardiac tissue, ATO led to the significant increase in cardiac lipid peroxidation (LPO) and nitric oxide (NO); remarkable decrease in the activity of cardiac antioxidant enzymes such as catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx); and the depletion of the total antioxidant capacity (TAC) and total thiol groups (TTGs). PTX was able to reduce the increased levels of serum cardiac markers (LDH, CPK, cTnI, TC, and TG), cardiac LPO, and improve antioxidant markers (TAC, TTGs, CAT, SOD, and GPx) alongside histopathologic changes. However, no significant changes were observed in elevated serum glucose and cardiac NO levels. In conclusion, the current study showed the potential therapeutic effect of PTX in the prevention of ATO-induced cardiotoxicity via reversing the oxidative stress.
Collapse
|
8
|
Hotz PW, Wiesnet M, Tascher G, Braun T, Müller S, Mendler L. Profiling the Murine SUMO Proteome in Response to Cardiac Ischemia and Reperfusion Injury. Molecules 2020; 25:E5571. [PMID: 33260959 PMCID: PMC7731038 DOI: 10.3390/molecules25235571] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/20/2020] [Accepted: 11/24/2020] [Indexed: 01/31/2023] Open
Abstract
SUMOylation is a reversible posttranslational modification pathway catalyzing the conjugation of small ubiquitin-related modifier (SUMO) proteins to lysine residues of distinct target proteins. SUMOylation modifies a wide variety of cellular regulators thereby affecting a multitude of key processes in a highly dynamic manner. The SUMOylation pathway displays a hallmark in cellular stress-adaption, such as heat or redox stress. It has been proposed that enhanced cellular SUMOylation protects the brain during ischemia, however, little is known about the specific regulation of the SUMO system and the potential target proteins during cardiac ischemia and reperfusion injury (I/R). By applying left anterior descending (LAD) coronary artery ligation and reperfusion in mice, we detect dynamic changes in the overall cellular SUMOylation pattern correlating with decreased SUMO deconjugase activity during I/R injury. Further, unbiased system-wide quantitative SUMO-proteomics identified a sub-group of SUMO targets exhibiting significant alterations in response to cardiac I/R. Notably, transcription factors that control hypoxia- and angiogenesis-related gene expression programs, exhibit altered SUMOylation during ischemic stress adaptation. Moreover, several components of the ubiquitin proteasome system undergo dynamic changes in SUMO conjugation during cardiac I/R suggesting an involvement of SUMO signaling in protein quality control and proteostasis in the ischemic heart. Altogether, our study reveals regulated candidate SUMO target proteins in the mouse heart, which might be important in coping with hypoxic/proteotoxic stress during cardiac I/R injury.
Collapse
Affiliation(s)
- Paul W. Hotz
- Institute of Biochemistry II, Goethe University Medical School, University Hospital Building 75, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (P.W.H.); (G.T.)
| | - Marion Wiesnet
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany; (M.W.); (T.B.)
| | - Georg Tascher
- Institute of Biochemistry II, Goethe University Medical School, University Hospital Building 75, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (P.W.H.); (G.T.)
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Ludwigstrasse 43, 61231 Bad Nauheim, Germany; (M.W.); (T.B.)
| | - Stefan Müller
- Institute of Biochemistry II, Goethe University Medical School, University Hospital Building 75, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (P.W.H.); (G.T.)
| | - Luca Mendler
- Institute of Biochemistry II, Goethe University Medical School, University Hospital Building 75, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (P.W.H.); (G.T.)
| |
Collapse
|
9
|
Li J, Thangaiyan R, Govindasamy K, Wei J. Anti-inflammatory and anti-apoptotic effect of zingiberene on isoproterenol-induced myocardial infarction in experimental animals. Hum Exp Toxicol 2020; 40:915-927. [PMID: 33242989 DOI: 10.1177/0960327120975131] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This study aimed to investigate the antihyperlipidemic and anti-inflammatory effect of zingiberene (ZBN) on isoproterenol-(ISO) induced myocardial infarction in rats. ZBN (10 mg/kg b.wt.) was orally administered to rats for 21 days and ISO (85 mg/kg b.wt.) was subcutaneously injected into the rats at 24 h intervals for the last 2 consecutive days. We observed increased serum creatine kinase, creatine kinase-MB, cardiac troponin T, and I levels in ISO-treated MI rats. Conversely, ZBN oral administration significantly prevented in cardiac marker enzyme activities in ISO-mediated rats. We also noticed that ZBN oral administration prevented ISO-induced expression of lipid peroxidative markers, total cholesterol, triglycerides, phospholipids, free fatty acids, very-low-density lipoprotein cholesterol (VLDL-C), low-density lipoprotein cholesterol (LDL-C) to the normal basal level. Furthermore, ZBN restored ISO-mediated antioxidant status, increased level of high-density lipoprotein cholesterol (HDL-C), and tissue phospholipids to the near-normal levels. Besides, ZBN pre-treatment significantly reduced the level of inflammatory markers (TNF-α, IL-6, NF-κB, and IL-1β) in ISO-induced MI in rats. We noticed that ZBN pretreatment inhibited the pro-apoptotic proteins Bax and cytochrome c and increased the Bcl-2 expression in ISO induced rats. The gene expression profiling by qRT-PCR array illustrates that ZBN treatment prevents the ISO mediated activation of cardiac markers, inflammatory, and fibrosis-related genes in the heart tissue. Taken together, pre-treatment with ZBN attenuated ISO-induced MI resolved exhibits the anti-inflammatory and antiapoptotic effect.
Collapse
Affiliation(s)
- Jianwei Li
- Department of Cardiology, Xi'an Fourth Hospital, Xi'an, Shaanxi, China
| | - Radhiga Thangaiyan
- Department of Biochemistry and Biotechnology, 29895Annamalai University, Annamalai Nagar, Tamil Nadu, India
| | - Kanimozhi Govindasamy
- Department of Biochemistry, Dharmapuram Gnanambigai Government Arts College for Women, Mayiladuthurai, Tamil Nadu, India
| | - Jianxia Wei
- Department of Cardiology, The Third Affiliated Hospital of Xi 'an Medical College, Xi'an, Shaanxi, China
| |
Collapse
|
10
|
Zhou C, Yu F, Zeng P, Zhang T, Huang H, Chen W, Wu B. Circadian sensitivity to the cardiac glycoside oleandrin is associated with diurnal intestinal P-glycoprotein expression. Biochem Pharmacol 2019; 169:113622. [PMID: 31472126 DOI: 10.1016/j.bcp.2019.08.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 08/26/2019] [Indexed: 12/21/2022]
Abstract
The cardiac glycoside oleandrin is a main active constituent of the botanical anti-cancer drug candidate PBI-05204, an extract of Nerium oleander. Here, we aimed to determine the circadian sensitivity of mice to oleandrin, and to investigate the role of intestinal P-gp in generating rhythmic drug toxicity. Toxicity and pharmacokinetic experiments were performed with wild-type, Bmal1iKO (intestine-specific Bmal1 knockout) and Bmal1fl/fl (control littermates of Bmal1iKO) mice. The cardiac toxicity (reflected by plasma CK-MB, LDH and cTn-I levels) varied significantly with the times of drug dosing in wild-type mice (a lower toxicity at ZT10 and more severe at ZT2/22). Dosing at ZT2 generated a higher drug exposure than ZT10, supporting a lower toxicity at ZT10. Intracellular accumulation of oleandrin (2.5-10 μM) was reduced in MDCKⅡ-MDR1 than in parental cells. MDR1 overexpression decreased the cell sensitivity to oleandrin toxicity. The net flux ratio (MDCKⅡ-MDR1 versus parental cells) was 2.9 for oleandrin. These data indicated oleandrin as a P-gp substrate. Both mdr1a mRNA and P-gp protein oscillated with the times of the day in small intestine of Bmal1fl/fl mice. Intestinal ablation of Bmal1 down-regulated mdr1a mRNA and P-gp protein, and abrogated their rhythms. Likewise, Bmal1 silencing led to down-regulated mdr1a mRNA and to a loss of its rhythmicity in serum-shocked CT26 cells. Based on luciferase reporter assays, Bmal1 regulated rhythmic mdr1a transcription through the clock output genes Hlf and E4bp4. Intestinal ablation of Bmal1 exacerbated oleandrin toxicity and enhanced drug exposure. Moreover, time dependency of toxicity and drug exposure were lost in Bmal1iKO mice. In conclusion, diurnal intestinal P-gp is a critical factor influencing daily oleandrin exposure and toxicity. Our findings have implications in minimizing oleandrin (and possibly Nerium oleander) toxicity and improving drug efficacy via dosing time optimization.
Collapse
Affiliation(s)
- Cui Zhou
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, Guangzhou, China; College of Chemistry and Biology Engineering, Yichun University, Jiangxi, China
| | - Fangjun Yu
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, Guangzhou, China; International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Peng Zeng
- School of Food and Pharmaceutical Engineering, Zhaoqing University, Zhaoqing, Guangdong, China
| | - Tianpeng Zhang
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, Guangzhou, China
| | - Hao Huang
- College of Chemistry and Biology Engineering, Yichun University, Jiangxi, China
| | - Wenying Chen
- The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Baojian Wu
- Research Center for Biopharmaceutics and Pharmacokinetics, College of Pharmacy, Jinan University, Guangzhou, China; International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development of Chinese Ministry of Education (MOE), College of Pharmacy, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
11
|
Catalpol pretreatment attenuates cardiac dysfunction following myocardial infarction in rats. Anatol J Cardiol 2019; 19:296-302. [PMID: 29724983 PMCID: PMC6280265 DOI: 10.14744/anatoljcardiol.2018.33230] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Objective: To investigate the effects and mechanisms of catalpol on cardiac function in rats with isoproterenol (ISO)-induced myocardial infarction (MI). Methods: Adult male Wistar rats were divided into four groups: control group, ISO group, catalpol (L, low dose) group, and catalpol (H, high dose) group. Isoproterenol (85 mg/kg) was injected subcutaneously for 2 consecutive days to induce experimental MI. At the end of experiment, the effects of catalpol on cardiac function; apelin levels; apoptosis index; apelin, APJ, Bcl-2, and Bax protein expression; and caspase-3/9 activities were investigated. Results: The rats in the ISO group showed lower left ventricular maximum rate of positive or negative pressure development (±LVdp/dtmax) and left ventricular end-systolic pressure (LVSP) and higher left ventricular end-diastolic pressure (LVEDP) than those in the control group, suggesting severe cardiac dysfunction. Interestingly, catalpol administration significantly ameliorated the ISO-induced cardiac dysfunction. The groups administered low and high dosages catalpol (5 and 10 mg/kg/day, respectively) showed higher ±LVdp/dtmax and LVSP and lower LVEDP than the group administered ISO alone. Catalpol markedly upregulated apelin levels in the plasma and myocardium. Further, catalpol increased the apelin and APJ expression levels in the myocardium of the ISO-treated rats. In addition, catalpol pretreatment inhibited cardiomyocyte apoptosis as indicated by a decrease in the TUNEL-positive cell percentage, alterations in the Bax and Bcl-2 expression levels, and a decline in caspase-3 and caspase-9 activities. Conclusion: Our results revealed that catalpol can improve cardiac function. Its protective effects may be linked to the enhancement of myocardium contractility, regulation of the apelin/APJ pathway, and inhibition of cardiomyocyte apoptosis.
Collapse
|
12
|
Dejea H, Garcia-Canadilla P, Cook AC, Guasch E, Zamora M, Crispi F, Stampanoni M, Bijnens B, Bonnin A. Comprehensive Analysis of Animal Models of Cardiovascular Disease using Multiscale X-Ray Phase Contrast Tomography. Sci Rep 2019; 9:6996. [PMID: 31061429 PMCID: PMC6502928 DOI: 10.1038/s41598-019-43407-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/23/2019] [Indexed: 01/02/2023] Open
Abstract
Cardiovascular diseases (CVDs) affect the myocardium and vasculature, inducing remodelling of the heart from cellular to whole organ level. To assess their impact at micro and macroscopic level, multi-resolution imaging techniques that provide high quality images without sample alteration and in 3D are necessary: requirements not fulfilled by most of current methods. In this paper, we take advantage of the non-destructive time-efficient 3D multiscale capabilities of synchrotron Propagation-based X-Ray Phase Contrast Imaging (PB-X-PCI) to study a wide range of cardiac tissue characteristics in one healthy and three different diseased rat models. With a dedicated image processing pipeline, PB-X-PCI images are analysed in order to show its capability to assess different cardiac tissue components at both macroscopic and microscopic levels. The presented technique evaluates in detail the overall cardiac morphology, myocyte aggregate orientation, vasculature changes, fibrosis formation and nearly single cell arrangement. Our results agree with conventional histology and literature. This study demonstrates that synchrotron PB-X-PCI, combined with image processing tools, is a powerful technique for multi-resolution structural investigation of the heart ex-vivo. Therefore, the proposed approach can improve the understanding of the multiscale remodelling processes occurring in CVDs, and the comprehensive and fast assessment of future interventional approaches.
Collapse
Affiliation(s)
- Hector Dejea
- Paul Scherrer Institut, Villigen PSI, Switzerland.
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland.
| | - Patricia Garcia-Canadilla
- PhySense, DTIC, Universitat Pompeu Fabra, Barcelona, Spain
- Institute of Cardiovascular Science, University College London, London, UK
| | - Andrew C Cook
- Institute of Cardiovascular Science, University College London, London, UK
| | - Eduard Guasch
- Arrhythmia Unit, Department of Cardiology, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red - Cardiovascular (CIBER-CV), Madrid, Spain
| | - Monica Zamora
- BCNatal, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Fatima Crispi
- BCNatal, Hospital Clínic and Hospital Sant Joan de Déu, Barcelona, Spain
- Centre for Biomedical Research on Rare Diseases (CIBER-ER), Hospital Clínic, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marco Stampanoni
- Paul Scherrer Institut, Villigen PSI, Switzerland
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Bart Bijnens
- PhySense, DTIC, Universitat Pompeu Fabra, Barcelona, Spain
- ICREA, Barcelona, Spain
| | - Anne Bonnin
- Paul Scherrer Institut, Villigen PSI, Switzerland
| |
Collapse
|
13
|
Neumann U, Derwenskus F, Gille A, Louis S, Schmid-Staiger U, Briviba K, Bischoff SC. Bioavailability and Safety of Nutrients from the Microalgae Chlorella vulgaris, Nannochloropsis oceanica and Phaeodactylum tricornutum in C57BL/6 Mice. Nutrients 2018; 10:nu10080965. [PMID: 30049974 PMCID: PMC6116023 DOI: 10.3390/nu10080965] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/18/2018] [Accepted: 07/23/2018] [Indexed: 12/20/2022] Open
Abstract
Microalgae are rich in macronutrients and therefore, they have been proposed as a potential future food source preserving natural resources. Here, we studied safety and bioavailability of algae nutrients in mice. Three microalgae species, Chlorella vulgaris, Nannochloropsis oceanica and Phaeodactylum tricornutum, were studied after ball mill disruption at different doses (5%, 15% and 25% dry weight) for 14 days. In response to all three algae diets, we observed a weight gain similar or superior to that in response to the control diet. No substantial differences in organ weights nor gut length occurred. Protein bioavailability from the algae diets did not differ from the control diet ranging from 58% to 77% apparent biological value. Fat absorption was lower for microalgae compared to soy oil in control diets, albeit still substantial. High liver eicosapentaenoic acid levels were measured following feeding with N. oceanica, the algae richest in omega-3 fatty acids. Neither histological nor serum analyses revealed any heart, kidney or liver toxicity induced by any of the algae diets. Algae-rich diets were thus well accepted, well tolerated and suitable for the maintenance of body weight and normal organ function. No toxicological effects were observed.
Collapse
Affiliation(s)
- Ulrike Neumann
- Institute of Clinical Nutrition, University of Hohenheim, Fruwirthstr. 12, 70593 Stuttgart, Germany.
| | - Felix Derwenskus
- Institute of Interfacial Process Engineering and Plasma Technology, University of Stuttgart, 70569 Stuttgart, Germany.
| | - Andrea Gille
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany.
| | - Sandrine Louis
- Institute of Clinical Nutrition, University of Hohenheim, Fruwirthstr. 12, 70593 Stuttgart, Germany.
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany.
| | - Ulrike Schmid-Staiger
- Fraunhofer Institute for Interfacial Engineering and Biotechnology, 70569 Stuttgart, Germany.
| | - Karlis Briviba
- Department of Physiology and Biochemistry of Nutrition, Max Rubner-Institut, 76131 Karlsruhe, Germany.
| | - Stephan C Bischoff
- Institute of Clinical Nutrition, University of Hohenheim, Fruwirthstr. 12, 70593 Stuttgart, Germany.
| |
Collapse
|
14
|
Kalkan F, Parlakpinar H, Disli OM, Tanriverdi LH, Ozhan O, Polat A, Cetin A, Vardi N, Otlu YO, Acet A. Protective and therapeutic effects of dexpanthenol on isoproterenol-induced cardiac damage in rats. J Cell Biochem 2018; 119:7479-7489. [PMID: 29775243 DOI: 10.1002/jcb.27058] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 04/23/2018] [Indexed: 01/15/2023]
Abstract
The purpose of the study was to explore the protective and therapeutic effects of dexpanthenol (DEX) on isoproterenol (ISO)-induced cardiac damage. Forty rats were distributed into four groups: group I (Control); group II (ISO); ISO (150 mg/kg/day) was given to rats once a day for 2 consecutive days with an interval of 24 h; group III (DEX+ISO): DEX (250 mg/kg) was applied 30 min before the first ISO administration and continued in the next two days after second ISO administration; group IV (ISO+DEX): After the ISO treatment at 1st and 2nd days, DEX was given at 3rd and 4th days. Rats were monitored for mean arterial blood pressure (BP), heart rate, oxygen saturation (%SO2 ), and electrocardiography (ECG). Heart tissue levels of malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPX), reduced glutathione (GSH), total oxidant status (TOS); total antioxidant capacity (TAC), oxidative stress index (OSI), and caspase-3 were determined. BP and SO2 values indicated a significant decrease in the ISO group. Also, T wave negativity was observed in 6 of 10 rats, SOD, CAT, and GPX levels were significantly lower in ISO group than control group. ISO administration increased TOS and OSI levels, whereas DEX treatment significantly reduced these parameters. Also, ISO-induced morphological alterations such as disorganization of cardiomyocytes, loss of myofibrils and cytoplasmic vacuolization whereas these histological damages were significantly decreased in ISO+DEX and DEX+ISO groups when compared to the ISO group. This study implies the cardioprotective effects of DEX on ISO-induced cardiotoxicity.
Collapse
Affiliation(s)
- Ferhat Kalkan
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Hakan Parlakpinar
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Olcay M Disli
- Department of Cardiovascular Surgery, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Lokman H Tanriverdi
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Onural Ozhan
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Alaaddin Polat
- Department of Physiology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Aslı Cetin
- Department of Histology and Embryology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Nigar Vardi
- Department of Histology and Embryology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Yılmaz O Otlu
- Department of Cardiology, Faculty of Medicine, Inonu University, Malatya, Turkey
| | - Ahmet Acet
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, Turkey
| |
Collapse
|
15
|
Mattes WB, Goodsaid F. Regulatory landscapes for biomarkers and diagnostic tests: Qualification, approval, and role in clinical practice. Exp Biol Med (Maywood) 2017; 243:256-261. [PMID: 29110507 DOI: 10.1177/1535370217739629] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
While the term 'biomarker' is relatively new, the concept is millennia old. However, with the introduction of new technologies to discover potential biomarkers comes the need to assess their utility and veracity for any given use. This is particularly true for the use of biomarkers to support regulatory decisions in medical product development. Hence the US Food and Drug Administration has developed processes for the qualification of biomarkers and other medical product development tools, processes that are underscored by recent legislation (i.e. the 21st Century Cures Act). In addition to these qualification processes, diagnostic tests that measure a biomarker may follow a process for regulatory decision through the processes that evaluate companion diagnostics. This mini-review provides an overview of these processes and their role in pharmaceutical development and clinical use. Impact statement This work summarizes very recent developments in the US FDA's biomarker qualification program. Furthermore, it contrasts biomarker qualification with companion diagnostic evaluation. As such, it will be highly informative for researches considering taking a biomarker discovery farther along the road to validation.
Collapse
Affiliation(s)
- William B Mattes
- 1 National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA
| | | |
Collapse
|
16
|
Tanriverdi LH, Parlakpinar H, Ozhan O, Ermis N, Polat A, Vardi N, Tanbek K, Yildiz A, Acet A. Inhibition of NADPH oxidase by apocynin promotes myocardial antioxidant response and prevents isoproterenol-induced myocardial oxidative stress in rats. Free Radic Res 2017; 51:772-786. [PMID: 28969461 DOI: 10.1080/10715762.2017.1375486] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Preventive and/or therapeutic interventions for ischemic heart disease have gained considerable attention worldwide. We investigated the mechanism(s) underlying cardioprotection of apocynin (APO) and whether it attenuates isoproterenol (ISO)-induced myocardial damage in vivo. Thirty-two male Wistar Albino rats were randomised into four groups (n = 8 for each group): Group I (Control); Group II (ISO), ISO was given intraperitoneally (ip) (150 mg/kg/d) daily for 2 consecutive days; Group III (APO + ISO), APO was applied ip 20 mg/kg 30 min before the first ISO administration and continued for the next 2 d after the second ISO administration; Group IV (ISO + APO), after the ISO treatment on days 1 and 2, 20 mg/kg APO was given ip on days 3 and 4. Cardioprotective effects of APO were evaluated by biochemical values, histopathological observations and the antiapoptotic relative proteins. Mean blood pressure, heart rate, and electrocardiography (ECG) were also monitored. Malondialdehyde (MDA), superoxide dismutase (SOD), catalase (CAT), reduced glutathione (GSH), total oxidant status (TOS), total antioxidant capacity (TAC), oxidative stress index (OSI), caspase-3 and connexin 43 levels were determined. Major ECG changes were observed in the ISO-treated rats. MDA, TOS, OSI and creatine kinase levels decreased and SOD, CAT, GSH and TAC levels increased, indicating that APO reduced cardiac injury and oxidative stress compared with controls. APO also decreased the number of cardiomyocytes with pyknotic nuclei, inflammatory cell infiltration, intracytoplasmic vacuolisation and myofibrils. APO provides preventive and therapeutic effects on ISO-induced myocardial injury in rats by inhibiting reactive oxygen species production, blocking inflammation and enhancing antioxidant status.
Collapse
Affiliation(s)
- Lokman H Tanriverdi
- a Department of Medical Pharmacology , Faculty of Medicine, Inonu University , Malatya , Turkey
| | - Hakan Parlakpinar
- a Department of Medical Pharmacology , Faculty of Medicine, Inonu University , Malatya , Turkey
| | - Onural Ozhan
- a Department of Medical Pharmacology , Faculty of Medicine, Inonu University , Malatya , Turkey
| | - Necip Ermis
- b Department of Cardiology , Faculty of Medicine, Inonu University , Malatya , Turkey
| | - Alaadin Polat
- c Department of Physiology , Faculty of Medicine, Inonu University , Malatya , Turkey
| | - Nigar Vardi
- d Department of Histology and Embryology , Faculty of Medicine, Inonu University , Malatya , Turkey
| | - Kevser Tanbek
- c Department of Physiology , Faculty of Medicine, Inonu University , Malatya , Turkey
| | - Azibe Yildiz
- d Department of Histology and Embryology , Faculty of Medicine, Inonu University , Malatya , Turkey
| | - Ahmet Acet
- a Department of Medical Pharmacology , Faculty of Medicine, Inonu University , Malatya , Turkey
| |
Collapse
|
17
|
Das M, Howell M, Foran EA, Iyre R, Mohapatra SS, Mohapatra S. Sertoli Cells Loaded with Doxorubicin in Lipid Micelles Reduced Tumor Burden and Dox-Induced Toxicity. Cell Transplant 2017; 26:1694-1702. [PMID: 29251108 PMCID: PMC5753976 DOI: 10.1177/0963689717721223] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 04/18/2017] [Accepted: 04/25/2017] [Indexed: 12/26/2022] Open
Abstract
The toxic side effects of doxorubicin (Dox) limit its long-term use as a lung cancer chemotherapeutic. Additionally, drug delivery to the deep lung is challenging. To address these challenges, isolated rat Sertoli cells (SCs) were preloaded with Dox conjugated to lipid micelle nanoparticles (SC-DLMNs) and delivered to mouse lungs. These immunocompetent cells, when injected intravenously, travel to the lung, deliver the payload, and get cleared by the system quickly without causing any adverse reaction. We observed that SC-DLMNs effectively treated Lewis lung carcinoma 1-induced lung tumors in mice and the drug efficacy was comparable to SC-Dox treatment. Mice treated with SC-DLMNs also showed significantly less toxicity compared to those treated with SC-Dox. The encapsulation of Dox in lipid micelle nanoparticles reduced the toxicity of Dox and the SC-based delivery method ensured drug delivery to the deep lung without evoking any immune response. Taken together, these results provide a novel SC-based nanoparticle drug delivery method for improved therapeutic outcome of cardiotoxic antilung cancer drugs.
Collapse
Affiliation(s)
- Mahasweta Das
- Center for Research and Education in Nanobioengineering, University of South Florida College of Medicine, Tampa, FL, USA
- Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL, USA
| | - Mark Howell
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, USA
| | - Elspeth A. Foran
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, USA
| | - Rohit Iyre
- Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL, USA
| | - Shyam S. Mohapatra
- Center for Research and Education in Nanobioengineering, University of South Florida College of Medicine, Tampa, FL, USA
- Department of Internal Medicine, University of South Florida College of Medicine, Tampa, FL, USA
- James A. Haley Veterans Hospital, Tampa, FL, USA
| | - Subhra Mohapatra
- Center for Research and Education in Nanobioengineering, University of South Florida College of Medicine, Tampa, FL, USA
- Department of Molecular Medicine, University of South Florida College of Medicine, Tampa, FL, USA
- James A. Haley Veterans Hospital, Tampa, FL, USA
| |
Collapse
|
18
|
Beger R, Yu LR, Daniels J, Mattes W. Exploratory biomarkers: Analytical approaches and their implications. CURRENT OPINION IN TOXICOLOGY 2017. [DOI: 10.1016/j.cotox.2017.06.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
19
|
Smith A, Calley J, Mathur S, Qian HR, Wu H, Farmen M, Caiment F, Bushel PR, Li J, Fisher C, Kirby P, Koenig E, Hall DG, Watson DE. The Rat microRNA body atlas; Evaluation of the microRNA content of rat organs through deep sequencing and characterization of pancreas enriched miRNAs as biomarkers of pancreatic toxicity in the rat and dog. BMC Genomics 2016; 17:694. [PMID: 27576563 PMCID: PMC5006322 DOI: 10.1186/s12864-016-2956-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 07/20/2016] [Indexed: 02/06/2023] Open
Abstract
Background MicroRNAs (miRNA) are ~19–25 nucleotide long RNA molecules that fine tune gene expression through the inhibition of translation or degradation of the mRNA through incorporation into the RNA induced silencing complex (RISC). MicroRNAs are stable in the serum and plasma, are detectable in a wide variety of body fluids, are conserved across veterinary species and humans and are expressed in a tissue specific manner. They can be detected at low concentrations in circulation in animals and humans, generating interest in the utilization of miRNAs as serum and/or plasma based biomarkers of tissue injury. MicroRNA tissue profiling in rodents has been published, but sample an insufficient number of organs of toxicologic interest using microarray or qPCR technologies for miRNA detection. Here we impart an improved rat microRNA body atlas consisting of 21 and 23 tissues of toxicologic interest from male and female Sprague Dawley rats respectively, using Illumina miRNA sequencing. Several of the authors created a dog miRNA body atlas and we collaborated to test miRNAs conserved in rat and dog pancreas in caerulein toxicity studies utilizing both species. Results A rich data set is presented that more robustly defines the tissue specificity and enrichment profiles of previously published and undiscovered rat miRNAs. We generated 1,927 sequences that mapped to mature miRNAs in rat, mouse and human from miRBase and discovered an additional 1,162 rat miRNAs as compared to the current number of rat miRNAs in miRBase version 21. Tissue specific and enriched miRNAs were identified and a subset of these miRNAs were validated by qPCR for tissue specificity or enrichment. As an example of the power of this approach, we have conducted rat and dog pancreas toxicity studies and examined the levels of some tissue specific and enriched miRNAs conserved between rat and dog in the serum of each species. The studies demonstrate that conserved tissue specific/enriched miRs-216a-5p, 375-3p, 148a-3p, 216b-5p and 141-3p are candidate biomarkers of pancreatic injury in the rat and dog. Conclusions A microRNA body atlas for rat and dog was useful in identifying new candidate miRNA biomarkers of organ toxicity in 2 toxicologically relevant species. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2956-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aaron Smith
- Department of Investigative Toxicology, Non Clinical Safety Assessment and Pathology, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, 46285, IN, USA.
| | - John Calley
- Department of TTX Bioinformatics, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, 46285, IN, USA
| | - Sachin Mathur
- Department of TTX Bioinformatics, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, 46285, IN, USA
| | - Hui-Rong Qian
- Department of Discovery and Development Statistics, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, 46285, IN, USA
| | - Han Wu
- Department of Discovery and Development Statistics, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, 46285, IN, USA
| | - Mark Farmen
- Department of Discovery and Development Statistics, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, 46285, IN, USA
| | - Florian Caiment
- Department of Toxicogenomics, Maastricht University, Universiteitsingel, Maastricht, The Netherlands
| | - Pierre R Bushel
- National Institute of Environmental Health Sciences, Biostatistics Branch, Durham, NC, USA
| | - Jianying Li
- Kelly Government Solutions, Research Triangle Park, Durham, NC, 27709, USA
| | - Craig Fisher
- Drug Safety Evaluation, Takeda Pharmaceuticals International Company, Deerfield, USA
| | - Patrick Kirby
- Drug Safety Evaluation, Takeda Pharmaceuticals International Company, Deerfield, USA
| | - Erik Koenig
- Molecular Pathology, Takeda Pharmaceuticals International Company, Deerfield, USA
| | - David G Hall
- Department of Investigative Pathology, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, 46285, IN, USA
| | - David E Watson
- Department of Investigative Toxicology, Non Clinical Safety Assessment and Pathology, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, 46285, IN, USA
| |
Collapse
|
20
|
Engle SK, Watson DE. Natriuretic Peptides as Cardiovascular Safety Biomarkers in Rats: Comparison With Blood Pressure, Heart Rate, and Heart Weight. Toxicol Sci 2015; 149:458-72. [PMID: 26609138 DOI: 10.1093/toxsci/kfv240] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular (CV) toxicity is an important cause of failure during drug development. Blood-based biomarkers can be used to detect CV toxicity during preclinical development and prioritize compounds at lower risk of causing such toxicities. Evidence of myocardial degeneration can be detected by measuring concentrations of biomarkers such as cardiac troponin I and creatine kinase in blood; however, detection of functional changes in the CV system, such as blood pressure, generally requires studies in animals with surgically implanted pressure transducers. This is a significant limitation because sustained changes in blood pressure are often accompanied by changes in heart rate and together can lead to cardiac hypertrophy and myocardial degeneration in animals, and major adverse cardiovascular events (MACE) in humans. Increased concentrations of NPs in blood correlate with higher risk of cardiac mortality, all-cause mortality, and MACE in humans. Their utility as biomarkers of CV function and toxicity in rodents was investigated by exploring the relationships between plasma concentrations of NTproANP and NTproBNP, blood pressure, heart rate, and heart weight in Sprague Dawley rats administered compounds that caused hypotension or hypertension, including nifedipine, fluprostenol, minoxidil, L-NAME, L-thyroxine, or sunitinib for 1-2 weeks. Changes in NTproANP and/or NTproBNP concentrations were inversely correlated with changes in blood pressure. NTproANP and NTproBNP concentrations were inconsistently correlated with relative heart weights. In addition, increased heart rate was associated with increased heart weights. These studies support the use of natriuretic peptides and heart rate to detect changes in blood pressure and cardiac hypertrophy in short-duration rat studies.
Collapse
Affiliation(s)
- Steven K Engle
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, Indiana 46285
| | - David E Watson
- Lilly Research Laboratories, Division of Eli Lilly and Company, Indianapolis, Indiana 46285
| |
Collapse
|
21
|
Frobert A, Valentin J, Magnin JL, Riedo E, Cook S, Giraud MN. Prognostic Value of Troponin I for Infarct Size to Improve Preclinical Myocardial Infarction Small Animal Models. Front Physiol 2015; 6:353. [PMID: 26640441 PMCID: PMC4661277 DOI: 10.3389/fphys.2015.00353] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 11/09/2015] [Indexed: 11/13/2022] Open
Abstract
UNLABELLED Coronary artery ligations to induce myocardial infarction (MI) in mice and rats are widely used in preclinical investigation. However, myocardial ischemic damage and subsequent infarct size are highly variable. The lack of standardization of the model impairs the probability of effective translation to the clinic. Cardiac Troponin I (cTnI) is a major clinically relevant biomarker. AIM In the present study, we investigated the prognostic value of cTnI for early estimation of the infarct size. METHODS AND RESULTS Infarcts of different sizes were induced in mice and rats by ligation, at a random site, of the coronary artery. Kinetics of the plasma levels of cTnI were measured. Heart function was evaluated by echocardiography, the percentage of infarcted left ventricle and infarct expansion index were assessed from histological section. We observed that plasma cTnI level peaked at 24 h in the infarcted rats and between 24 and 48 h in mice. Sham operated animals had a level of cTnI below 15 ng/mL. Infarct expansion index (EI) assessed 4 weeks after ligation showed a large variation coefficient of 63 and 71% in rats and mice respectively. We showed a significative correlation between cTnI level and the EI demonstrating its predictive value for myocardial injury in small animal models. CONCLUSION we demonstrated the importance of cTnI plasma level as a major early marker to assist in the optimal and efficient management of MI in laboratory animals model. The presented results stress the need for comparable biomarkers in the animal model and clinical trials for improved translation.
Collapse
Affiliation(s)
- Aurélien Frobert
- Cardiology, Department of Medicine, University and Hospital of Fribourg Fribourg, Switzerland
| | - Jérémy Valentin
- Cardiology, Department of Medicine, University and Hospital of Fribourg Fribourg, Switzerland
| | | | - Erwin Riedo
- Central Laboratory, Hospital Fribourg Fribourg, Switzerland
| | - Stéphane Cook
- Cardiology, Department of Medicine, University and Hospital of Fribourg Fribourg, Switzerland
| | - Marie-Noëlle Giraud
- Cardiology, Department of Medicine, University and Hospital of Fribourg Fribourg, Switzerland
| |
Collapse
|
22
|
Maher TJ, Ren Y, Li Q, Braunlin E, Garry MG, Sorrentino BP, Martin CM. ATP-binding cassette transporter Abcg2 lineage contributes to the cardiac vasculature after oxidative stress. Am J Physiol Heart Circ Physiol 2014; 306:H1610-8. [PMID: 24727496 DOI: 10.1152/ajpheart.00638.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Due to their specialized location, stem and progenitor cells are often exposed to oxidative stress. Although ATP-binding cassette transporter subfamily G member 2 (Abcg2)-expressing cells have been implicated in cardiac protective mechanisms involving oxidative stress, there remains a lack of understanding regarding the behavior of cardiac Abcg2-expressing cells when exposed to ROS. The aim of the present study was to characterize the response of the cardiac Abcg2 lineage to oxidative stress. In vitro analysis demonstrated that the antioxidant program regulated by Abcg2 is dependent on a functional transporter. Delivery of paraquat dichloride (PQ), a systemic oxidative stress-inducing agent, to mice confirmed that Abcg2 provides a survival benefit. When exposed to PQ, reporter mice showed an increase in the Abcg2 lineage. Transcriptional and immunohistochemical analysis of Abcg2 lineage-positive cells revealed an enhanced vascular commitment after stress. Finally, preconditioning with PQ demonstrated a reduction in scar size and an increase in angiogenesis after permanent left coronary artery ligation. In conclusion, the data suggest that Abcg2 plays a cytoprotective role in response to in vivo oxidative stress. The contribution of the Abcg2 lineage to the vasculature in the heart is increased after PQ delivery.
Collapse
Affiliation(s)
| | - Yi Ren
- University of Minnesota, Minneapolis, Minnesota; and
| | - Qinglu Li
- University of Minnesota, Minneapolis, Minnesota; and
| | | | - Mary G Garry
- University of Minnesota, Minneapolis, Minnesota; and
| | | | | |
Collapse
|
23
|
Liu L, Aguirre SA, Evering WEN, Hirakawa BP, May JR, Palacio K, Wang J, Zhang Y, Stevens GJ. miR-208a as a Biomarker of Isoproterenol-induced Cardiac Injury in Sod2+/− and C57BL/6J Wild-type Mice. Toxicol Pathol 2014; 42:1117-29. [DOI: 10.1177/0192623314525684] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This investigation examined microRNA-208a (miR-208a) as a potential biomarker of isoproterenol (ISO)-induced cardiac injury in superoxide dismutase-2 ( Sod2+/−) and the wild-type mice, and the potential sensitivity of Sod2+/− mice to ISO-induced toxicity. A single intraperitoneal injection of ISO was administered to age-matched wild-type and Sod2+/− mice at 0, 80, or 160 mg/kg. Plasma miR-208a, cardiac troponin I (cTnI), and ISO systemic exposure were measured at various time points postdose. Hearts were collected for histopathology examination and for tissue expression of miR-208a and myosin heavy chain 7. ISO administration caused increases in cTnI and miR-208a plasma levels that correlated with myocardial damage; however, the magnitude of increase differed according to the types of mice. At similar ISO systemic exposure, the magnitude of cTnI was greater in wild-type mice compared to Sod2+/− mice; however, the magnitude of miR-208a was greater in Sod2+/− mice than that of the wild-type mice. Myocardial degeneration occurred at ≥3 hr in the wild-type and ≥6 hr in Sod2+/− mice. At ≥24 hr after ISO administration, miR-208a appeared superior to cTnI in indicating myocardial injury in both wild-type and Sod2+/− mice. Sod2+/− mice were not more sensitive than wild-type mice to ISO-induced toxicity.
Collapse
Affiliation(s)
- Ling Liu
- Pfizer Global Research and Development, Drug Safety Research and Development, San Diego, California, USA
| | - Shirley A. Aguirre
- Pfizer Global Research and Development, Drug Safety Research and Development, San Diego, California, USA
| | - Winston E. N. Evering
- Pfizer Global Research and Development, Drug Safety Research and Development, San Diego, California, USA
| | - Brad P. Hirakawa
- Pfizer Global Research and Development, Drug Safety Research and Development, San Diego, California, USA
| | - Jeffrey R. May
- Pfizer Global Research and Development, Drug Safety Research and Development, San Diego, California, USA
| | - Kimbie Palacio
- Pfizer Global Research and Development, Drug Safety Research and Development, San Diego, California, USA
| | - Jianying Wang
- Pfizer Global Research and Development, Drug Safety Research and Development, San Diego, California, USA
| | - Yizhong Zhang
- Pfizer Global Research and Development, Bioanalytical Research, Groton, Connecticut, USA
| | - Gregory J. Stevens
- Pfizer Global Research and Development, Drug Safety Research and Development, San Diego, California, USA
| |
Collapse
|
24
|
Evran B, Karpuzoğlu H, Develi S, Kalaz EB, Soluk-Tekkeşin M, Olgaç V, Doğru-Abbasoğlu S, Uysal M. Effects of carnosine on prooxidant–antioxidant status in heart tissue, plasma and erythrocytes of rats with isoproterenol-induced myocardial infarction. Pharmacol Rep 2014; 66:81-6. [DOI: 10.1016/j.pharep.2013.08.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 07/22/2013] [Accepted: 08/13/2013] [Indexed: 10/25/2022]
|
25
|
Hausner EA, Hicks KA, Leighton JK, Szarfman A, Thompson AM, Harlow P. Qualification of cardiac troponins for nonclinical use: a regulatory perspective. Regul Toxicol Pharmacol 2013; 67:108-14. [PMID: 23876748 DOI: 10.1016/j.yrtph.2013.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 07/10/2013] [Accepted: 07/13/2013] [Indexed: 01/25/2023]
Abstract
The US Food and Drug Administration (FDA) Biomarker Qualification Review Team presents its perspective on the recent qualification of cardiac troponins for use in nonclinical safety assessment studies. The goal of this manuscript is to provide greater transparency into the qualification process and factors that were considered in reaching a regulatory decision. This manuscript includes an overview of the data that were submitted and a discussion of the strengths and shortcomings of these data supporting the qualification decision. The cardiac troponin submission is the first literature-based biomarker application to be reviewed by the FDA and insights gained from this experience may aid future submissions and help streamline the characterization and qualification of future biomarkers.
Collapse
Affiliation(s)
- E A Hausner
- Division of Cardiovascular and Renal Products, Center for Drug Evaluation and Research, Office of New Drugs, US Food and Drug Administration, United States.
| | | | | | | | | | | |
Collapse
|
26
|
Tonomura Y, Matsushima S, Kashiwagi E, Fujisawa K, Takagi S, Nishimura Y, Fukushima R, Torii M, Matsubara M. Biomarker panel of cardiac and skeletal muscle troponins, fatty acid binding protein 3 and myosin light chain 3 for the accurate diagnosis of cardiotoxicity and musculoskeletal toxicity in rats. Toxicology 2012; 302:179-89. [PMID: 22878004 DOI: 10.1016/j.tox.2012.07.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Revised: 07/24/2012] [Accepted: 07/25/2012] [Indexed: 11/17/2022]
Abstract
Cardiotoxicity and musculoskeletal toxicity can be life-threatening, and thus have strong impact on both the development and marketing of drugs. Because the conventional biomarkers such as aspartate aminotransferase (AST), lactate dehydrogenase (LDH), and creatine kinase (CK) have low detection power, there has been increasing interest in developing biomarkers with higher detection power. The current study examined the usefulness of several promising biomarkers, cardiac and skeletal muscle troponins (cTnI, cTnT and sTnI), fatty acid binding protein 3 (FABP3) and myosin light chain 3 (MYL3), and compared the obtained data to AST, LDH and CK in rat models treated with various myotoxic and non-myotoxic compounds (isoproterenol, metaproterenol, doxorubicin, mitoxantrone, allylamine, cyclosporine A, cyclophosphamide, aminoglutethimide, acetaminophen, methapyrilene, allylalcohol and α-naphthylisothiocyanate). These promising biomarkers were found to be superior to the conventional biomarkers, as they had a specific and abundant distribution within the heart and/or skeletal muscles; exhibited a positive correlation between the amplitude of increases and the degree of pathological alterations; had higher diagnostic accuracy for detecting pathological alterations; and had the additive effect of improving the diagnostic accuracy of conventional biomarkers. However, these promising biomarkers have several drawbacks including a rapid clearance, the fact that they are affected by renal dysfunction, and different reactivity to the mode of action of individual myotoxicants. In conclusion, the promising biomarkers cTnI, cTnT, FABP3, MYL3, and sTnI demonstrated sensitivity and specificity for cardiac and skeletal myotoxicity that was superior to those of conventional biomarkers, while we should pay attention to the drawbacks of these biomarkers when used in toxicity studies.
Collapse
Affiliation(s)
- Yutaka Tonomura
- Drug Safety Evaluation, Drug Developmental Research Laboratories, Shionogi & Co., Ltd., Osaka, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Mikaelian I, Scicchitano M, Mendes O, Thomas RA, Leroy BE. Frontiers in preclinical safety biomarkers: microRNAs and messenger RNAs. Toxicol Pathol 2012; 41:18-31. [PMID: 22659243 DOI: 10.1177/0192623312448939] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The measurement of plasma microRNAs (miRNAs) and messenger RNAs (mRNAs) is the most recent effort to identify novel biomarkers in preclinical safety. These genomic markers often display tissue-specific expression, may be released from the tissues into the plasma during toxic events, change early and with high magnitude in tissues and in the blood during specific organ toxicities, and can be measured using multiplex formats. Their validation as biomarkers has been challenged by the technical difficulties. In particular, the concentration of miRNAs in the plasma depends on contamination by miRNAs originating from blood cells and platelets, and the relative fraction of miRNAs in complexes with Argonaute 2, high-density lipoproteins, and in exosomes and microvesicles. In spite of these hurdles, considerable progress has recently been made in assessing the potential value of miRNAs in the clinic, especially in cancer patients and cardiovascular diseases. The future of miRNAs and mRNAs as biomarkers of disease and organ toxicity depends on our ability to characterize their kinetics and to establish robust collection and measurement methods. This review covers the basic biology of miRNAs and the published literature on the use of miRNAs and mRNAs as biomarkers of specific target organ toxicity.
Collapse
|
28
|
Ojha S, Golechha M, Kumari S, Arya DS. Protective effect of Emblica officinalis (amla) on isoproterenol-induced cardiotoxicity in rats. Toxicol Ind Health 2011; 28:399-411. [PMID: 22033422 DOI: 10.1177/0748233711413798] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Emblica officinalis, commonly known as amla, is an important medicinal plant reputed for its dietary and therapeutic uses. The aim of the present study was to investigate the protective role of E. officinalis against isoproterenol (ISP)-induced cardiotoxicity in rats and elucidate the possible mechanism involved. Rats were administered E. officinalis (100, 250 and 500 mg/kg, p.o.) or vehicle (normal saline) for 30 days, with concurrent subcutaneous injections of ISP (85 mg/kg, at 24 h interval) on 29th and 30th day. ISP-induced cardiac dysfunction as evidenced by decreased mean arterial pressure, heart rate, contractility (+LVdP/dt) and relaxation (-LVdP/dt) along with increased left ventricular end diastolic pressure. ISP significantly (p < 0.05) decreased antioxidant enzymes, superoxide dismutase, catalase and glutathione peroxidase and myocyte-injury-specific marker enzymes, creatine phosphokinase-MB and lactate dehydrogenase in heart. A significant (p < 0.05) depletion of reduced glutathione and increase in thiobarbituric acid reactive substances along with histopathological alteration has further indicated the oxidative damage of myocardium. However, pretreatment with E. officinalis exhibited restoration of hemodynamic and left ventricular function along with significant preservation of antioxidants, myocytes-injury-specific marker enzymes and significant inhibition of lipid peroxidation. Furthermore, histopathological salvage of myocardium reconfirmed the protective effects of E. officinalis. Results of the present study demonstrate cardioprotective potential of E. officinalis attributed to its potent antioxidant and free radical scavenging activity as evidenced by favorable improvement in hemodynamic, contractile function and tissue antioxidant status.
Collapse
Affiliation(s)
- Shreesh Ojha
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | | | | | | |
Collapse
|
29
|
Assessment of subclinical, toxicant-induced hepatic gene expression profiles after low-dose, short-term exposures in mice. Regul Toxicol Pharmacol 2011; 60:54-72. [DOI: 10.1016/j.yrtph.2011.02.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 02/09/2011] [Accepted: 02/09/2011] [Indexed: 12/19/2022]
|
30
|
Abstract
Cardiac troponin (cTn) is a sensitive and specific biomarker for assessing cardiac damage and should be utilized in drug safety assessment. Lactate dehydrogenase and creatine kinase isoenzyme analyses have historically been used in pre-clinical toxicity testing to assess cardiac injury, but since these assays are less sensitive and specific than cTn, isoenzyme analyses, as determined by the manual electrophoretic technique, are no longer warranted. Commercial cTn assays developed for humans do not have the same immunoreactivity and functional sensitivity in the common pre-clinical testing species, so it is important to show that the assay that is chosen is appropriate for the pre-clinical species being assessed. The kinetics of the cTn response depends on the dose and frequency of test article administration as well as the mechanism of the cardiac injury induced by the test article. Cardiac troponin should be used in the assessment of classes of compound that have previously been shown to induce cardiac necrosis or if cardiac necrosis is identified histologically with a novel compound. Next generation high sensitivity cTn assays are being developed and the low levels of cTn detected with these assays may be an early sign of possibly reversible damage to the heart.
Collapse
Affiliation(s)
- William J. Reagan
- Drug Safety Research and Development, Pfizer Global Research and Development, Groton, Connecticut, USA
| |
Collapse
|