1
|
Awais M, Zubair HM, Nadeem H, Hill JW, Ali J, Saleem A, Asghar R, Khan S, Maqbool T, Akhtar MF, Naveed M, Asif M. Benzimidazole Derivative (N-{4-[2-(4-Methoxyphenyl)-1H-Benzimidazole-1-Sulfonyl] Phenyl} Acetamide) Ameliorates Methotrexate-Induced Intestinal Mucositis by Suppressing Oxidative Stress and Inflammatory Markers in Mice. Inflammation 2024; 47:1185-1203. [PMID: 38289578 DOI: 10.1007/s10753-024-01969-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 01/05/2024] [Accepted: 01/05/2024] [Indexed: 08/24/2024]
Abstract
Methotrexate (MTX)-induced intestinal mucositis (IM) is a common side effect in cancer treatment that impairs the immune system and gut microbes, resulting in loss of mucosal integrity and gut barrier dysfunction. The quality of life and outcomes of treatment are compromised by IM. The present study was designed to investigate the mucoprotective potential of the benzimidazole derivative N-{4-[2-(4-methoxyphenyl)-1H-benzimidazole-1-sulfonyl] phenyl} acetamide (B8) on MTX-induced IM in mice. IM was induced by a single dose of MTX in mice and assessed by physical manifestations as well as biochemical, oxidative, histological, and inflammatory parameters. B8 (1, 3, 9 mg/kg) significantly reduced diarrhea score, mitigated weight loss, increased feed intake and, survival rate in a dose-dependent manner. Notably, B8 exhibited a mucoprotective effect evident through the mitigation of villus atrophy, crypt hypoplasia, diminished crypt mitotic figures, mucin depletion, and oxidative stress markers (GSH, SOD, MDA, and catalase concentration). Gene expression analysis revealed that B8 downregulated the mRNA expression of tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (COX-2), interleukin-6 (IL-6), IL-1β, and nuclear factor-κB (NF-κB) and concurrently upregulated IL-10 expression in contrast to the MTX group. Further, B8 significantly improved the luminal microflora profile by augmenting the growth of Lactobacillus spp. and reducing the number of pathogenic bacteria (E. coli). Additionally, the enzyme-linked immunoassay showed that B8 decreased the levels of pro-inflammatory cytokines. Our findings suggest that B8 had mucoprotective effects against MTX-induced IM and could be used as an adjunct in chemotherapy to deter this side effect.
Collapse
Affiliation(s)
- Muhammad Awais
- Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Hafiz Muhammad Zubair
- Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan.
- Post-Graduate Medical College, Faculty of Medicine and Allied Health Sciences, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan.
| | - Humaira Nadeem
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, 43614, USA
| | - Jawad Ali
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, 38000, Pakistan
| | - Rabia Asghar
- Faculty of Pharmacy, The University of Lahore, Lahore, Pakistan
| | - Samiullah Khan
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Tahir Maqbool
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore, Pakistan
| | - Muhammad Naveed
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, 43614, USA
| | - Muhammad Asif
- Department of Pharmacology, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, 63100, Pakistan
| |
Collapse
|
2
|
Hassanein EHM, Ali FEM, Sayed MM, Mahmoud AR, Jaber FA, Kotob MH, Abd-Elhamid TH. Umbelliferone potentiates intestinal protective effect of Lactobacillus Acidophilus against methotrexate-induced intestinal injury: Biochemical and histological study. Tissue Cell 2023; 82:102103. [PMID: 37178526 DOI: 10.1016/j.tice.2023.102103] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 04/17/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023]
Abstract
Intestinal injury is a common adverse effect of methotrexate (MTX) therapy, limiting its clinical use. Despite oxidative stress and inflammation being the most embedded mechanism of injury, pharmacological agents that exhibit antioxidant and anti-inflammatory impacts could prevent such toxicities. This study aimed to assess the enteroprotective effect of lactobacillus acidophilus (LB) and/or umbelliferone (UMB) against MTX-induced intestinal injury. Histologically, pretreatment with LB, UMB, or their combinations preserve the intestinal histological structure and mucin content with superior effect in combination therapy. In addition, oral pretreatment with UMB, LB, or their combinations significantly restored oxidant/antioxidant status, as evidenced by the upregulation of Nrf2, SOD3, HO-1, GSH, and GST levels concurrent with a decline in MDA contents. Besides, they suppressed the inflammatory burden by inhibiting STAT3, MPO, TLR4, NF-κB, TNF-α, and IL-6 levels. Moreover, LB, UMB, or their combinations significantly upregulated Wnt and β-catenin expression. Notably, pretreatment with the combination therapy is superior to monotherapy in protecting rats' small intestines from MTX-induced enteritis. In conclusion, combined pretreatment with LB and UMB could be a novel therapeutic regimen for conditions of intestinal injury induced by MTX via restoring oxidant/antioxidant balance and suppressing inflammatory burden.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt
| | - Fares E M Ali
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt.
| | - Manal M Sayed
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Amany Refaat Mahmoud
- Department of Human Anatomy and Embryology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt; Department of Basic Medical Sciences, Unaizah College of Medicine and Medical Sciences, Qassim University, Unaizah, Kingdom of Saudi Arabia
| | - Fatima A Jaber
- Department of Biology, College of Science, University of Jeddah, P.O. Box 80327, Jeddah 21589, Saudi Arabia
| | - Mohamed H Kotob
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Tarek Hamdy Abd-Elhamid
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt; Department of Basic Medical Sciences, Faculty of Medicine, Aqaba Medical Sciences University, Aqaba 77110, Jordan
| |
Collapse
|
3
|
Armağan İ, Aşcı H, Erzurumlu Y, Özkula S, Hasseyid N, Kumbul Doğuç D, Okuyucu G, Varel A. Ramelteon and mechanism of its restorative effect in an experimental lung disease model. Toxicol Mech Methods 2023; 33:239-247. [PMID: 36482745 DOI: 10.1080/15376516.2022.2156006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Methotrexate (MTX) is an anticancer agent widely used in clinical practice for various oncological, rheumatological, autoimmune, and inflammatory diseases. However, the side effects of MTX limit its usage for treatment. In addition, diffuse alveolar damage, interstitial pneumonia, fibrosis, and pleural reactions may be encountered in MTX-induced pulmonary toxicity. Ramelteon (RML), a melatonin receptor agonist, has antioxidant, anti-inflammatory, and protective effects are shown by several studies. This study aimed to show the antioxidant, anti-inflammatory, and antiapoptotic effects of RML and its effect on the airway surface liquid volume homeostasis via aquaporins (AQP) in MTX-induced lung injury. Thirty-two female Wistar Albino rats were grouped into four groups as control, MTX (20 mg/kg, intraperitoneally, a single dose), MTX + RML, and RML (10 mg/kg, via oral gavage, for seven days) groups. Once the experiment ended, the rats' lung tissues were taken for biochemical, genetic, histopathological, and immunohistochemical examinations. MTX significantly increased oxidative stress index and total oxidative status, and decreased total antioxidant status levels by 202.0%, 141.4%, 20.2%, respectively, relative to the control (p ˂ 0.001 for all). AQP-1/5, which is an indicator of lung damage, was also found to decrease significantly (p ˂ 0.001). In addition, a significant increase was observed in interleukin-1β, interferon-beta, and caspase-8 expressions and histopathological changes as a result of immunohistochemical and histochemical examinations (p ˂ 0.001). RML treatment ameliorated all these changes and significantly regressed lung damage. Our results suggest that RML might be used as a lung-protective agent in various models of lung and tissue injury.
Collapse
Affiliation(s)
- İlkay Armağan
- Department of Histology and Embryology, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey
| | - Halil Aşcı
- Department of Pharmacology, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey
| | - Yalçın Erzurumlu
- Department of Biochemistry, Faculty of Pharmacy, Süleyman Demirel University, Isparta, Turkey
| | - Songül Özkula
- Department of Pharmacology, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey
| | - Nursel Hasseyid
- Department of Pharmacology, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey
| | - Duygu Kumbul Doğuç
- Department of Biochemistry, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey
| | - Gözde Okuyucu
- Department of Pathology, Faculty of Veterinary Medicine, Burdur Mehmet Akif Ersoy University, Burdur, Turkey
| | - Ahmetcan Varel
- Department of Histology and Embryology, Faculty of Medicine, Süleyman Demirel University, Isparta, Turkey
| |
Collapse
|
4
|
Hassanein EHM, Althagafy HS, Atwa AM, Kozman MR, Kotb El-Sayed MI, Soubh AA. Taurine attenuated methotrexate-induced intestinal injury by regulating NF-κB/iNOS and Keap1/Nrf2/HO-1 signals. Life Sci 2022; 311:121180. [PMID: 36370869 DOI: 10.1016/j.lfs.2022.121180] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/28/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022]
Abstract
Methotrexate (MTX) is a well-known and widely used cytotoxic chemotherapeutic agent. However, intestinal mucosa damage is a serious adverse effect of MTX. Taurine (TUR) is a sulfur-containing free β-amino acid with antioxidant and therapeutic value against several diseases. The current study aimed to determine the protective effect of TUR against MTX-induced intestinal injury. Rats were allocated into four groups. The first group received vehicles only. The second group received TUR at a dose of 250 mg/kg i.p. For induction of intestinal injury, the rats in the third group were given MTX once at a dose of 20 mg/kg, i.p. The fourth group received TUR 7 days before and 7 days after MTX, as previously described. TUR significantly attenuated the cytokine release by suppressing NF-κB and iNOS expressions. Moreover, cotreatment with TUR attenuated the increased MDA level while it enhanced the antioxidant GSH and SOD levels mediated by effective downregulation of Keap1 expression, while the expression of Nrf2, HO-1, and cytoglobin were up-regulated. Additionally, TUR mitigated the apoptosis and proliferation indices by decreasing the elevated levels of intestinal PCNA and caspase-3. Finally, TUR potently increased the cytotoxic activity of MTX toward Caco-2, MCF-7, and A549 cancer cells. In conclusion, TUR was a promising agent for relieving MTX-mediated intestinal injury via various antioxidant, anti-inflammatory, and antiapoptotic mechanisms.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut 71524, Egypt.
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Magy R Kozman
- Department of Clinical Pharmacology, Faculty of Pharmacy, Misr University for Science and Technology, Cairo, Egypt
| | - Mohamed I Kotb El-Sayed
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Ain Helwan, Helwan, Cairo, Egypt
| | - Ayman A Soubh
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, Giza 12566, Egypt
| |
Collapse
|
5
|
Kim H, Back JH, Han G, Lee SJ, Park YE, Gu MB, Yang Y, Lee JE, Kim SH. Extracellular vesicle-guided in situ reprogramming of synovial macrophages for the treatment of rheumatoid arthritis. Biomaterials 2022; 286:121578. [PMID: 35594838 DOI: 10.1016/j.biomaterials.2022.121578] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 05/02/2022] [Accepted: 05/11/2022] [Indexed: 12/01/2022]
Abstract
Activation state of synovial macrophages is significantly correlated with disease activity and severity of rheumatoid arthritis (RA) and provides valuable clues for RA treatment. Classically activated M1 macrophages in inflamed synovial joints secrete high levels of pro-inflammatory cytokines and chemokines, resulting in bone erosion and cartilage degradation. Herein, we propose extracellular vesicle (EV)-guided in situ macrophage reprogramming toward anti-inflammatory M2 macrophages as a novel RA treatment modality based on the immunotherapeutic concept of reestablishing M1-M2 macrophage equilibrium in synovial tissue. M2 macrophage-derived EVs (M2-EVs) were able to convert activated M1 into reprogrammed M2 (RM2) macrophages with extremely high efficiency (>90%), producing a distinct protein expression pattern characteristic of anti-inflammatory M2 macrophages. In particular, M2-EVs were enriched for proteins known to be involved in the generation and migration of M2 macrophages as well as macrophage reprogramming factors, allowing for rapid and efficient driving of macrophage polarization toward M2 phenotype. After administration of M2-EVs into the joint of a collagen-induced arthritis mouse model, the synovial macrophage polarization was significantly shifted from M1 to M2 phenotype, a process that benefited greatly from the long residence time (>3 days) of M2-EVs in the joint. This superb in situ macrophage-reprogramming ability of EVs resulted in decreased joint swelling, arthritic index score and synovial inflammation, with corresponding reductions in bone erosion and articular cartilage damage and no systemic toxicity. The anti-RA effects of M2-EVs were comparable to those of the conventional disease-modifying antirheumatic drug, Methotrexate, which causes a range of toxic adverse effects, including gastrointestinal mucosal injury. Overall, our EV-guided reprogramming strategy for in situ tuning of macrophage responses holds great promise for the development of anti-inflammatory therapeutics for the treatment of various inflammatory diseases in addition to RA.
Collapse
Affiliation(s)
- Hyosuk Kim
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Ji Hyun Back
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea; Chemical & Biological integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Geonhee Han
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Su Jin Lee
- Chemical & Biological integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Yae Eun Park
- Chemical & Biological integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Man Bock Gu
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Yoosoo Yang
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Ji Eun Lee
- Chemical & Biological integrative Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| | - Sun Hwa Kim
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
| |
Collapse
|
6
|
Konsman JP, Laaker CJ, Lloyd KR, Hiltz A, Smith BL, Smail MA, Reyes TM. Translationally relevant mouse model of early life cancer and chemotherapy exposure results in brain and small intestine cytokine responses: A potential link to cognitive deficits. Brain Behav Immun 2022; 99:192-202. [PMID: 34655730 PMCID: PMC8842482 DOI: 10.1016/j.bbi.2021.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 09/03/2021] [Accepted: 10/11/2021] [Indexed: 01/03/2023] Open
Abstract
Survivors of acute lymphoblastic leukemia (ALL), the most common childhood cancer, are at increased risk for long-term cognitive problems, including executive function deficits. The chemotherapeutic agent methotrexate (MTX) is used to treat most ALL patients and is closely associated with cognitive deficits. To address how early life cancer chemotherapy leads to cognitive deficits, we developed a translationally relevant mouse model of leukemia survival that exposed mice to leukemic cells and chemotherapeutic drugs (vincristine and MTX, with leucovorin rescue) in early life. Male and female mice were tested several weeks later using novel object recognition (recognition memory) and 5-choice serial reaction time task (executive function). Gene expression of proinflammatory, white matter and synapse-associated molecules was assessed in the prefrontal cortex and small intestine both acutely after chemotherapy and chronically after cognitive testing. Early life cancer-chemotherapy exposure resulted in recognition memory and executive function deficits in adult male mice. Prefrontal cortex expression of the chemokine Ccl2 was increased acutely, while small intestine expression of the proinflammatory cytokine tumor necrosis factor-alpha was elevated both acutely (both sexes) and chronically (males only). Inflammation in the small intestine was correlated with prefrontal cortical proinflammatory and synaptic gene expression changes, as well as to executive function deficits. Collectively, these data indicate that the current protocol results in a robust mouse model in which to study cognitive deficits in leukemia survivors, and suggest that small intestine inflammation may represent a novel contributor to adverse CNS consequences of early life chemotherapy.
Collapse
Affiliation(s)
- Jan Pieter Konsman
- Aquitaine Institute for Integrative and Cognitive Neuroscience (INCIA) UMR CNRS 5287, University of Bordeaux, 33076 Bordeaux, France
| | - Collin J. Laaker
- University of Cincinnati, College of Medicine, Department of Pharmacology and Systems Physiology, Cincinnati, OH, USA
| | - Kelsey R. Lloyd
- University of Cincinnati, College of Medicine, Department of Pharmacology and Systems Physiology, Cincinnati, OH, USA
| | - Adam Hiltz
- University of Cincinnati, College of Medicine, Department of Pharmacology and Systems Physiology, Cincinnati, OH, USA
| | - Brittany L. Smith
- University of Cincinnati, College of Medicine, Department of Pharmacology and Systems Physiology, Cincinnati, OH, USA
| | - Marissa A. Smail
- University of Cincinnati, College of Medicine, Department of Pharmacology and Systems Physiology, Cincinnati, OH, USA
| | - Teresa M. Reyes
- University of Cincinnati, College of Medicine, Department of Pharmacology and Systems Physiology, Cincinnati, OH, USA
| |
Collapse
|
7
|
Utilizing Melatonin to Alleviate Side Effects of Chemotherapy: A Potentially Good Partner for Treating Cancer with Ageing. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6841581. [PMID: 32566095 PMCID: PMC7260648 DOI: 10.1155/2020/6841581] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 05/14/2019] [Accepted: 05/27/2019] [Indexed: 02/07/2023]
Abstract
Persistent senescence seems to exert detrimental effects fostering ageing and age-related disorders, such as cancer. Chemotherapy is one of the most valuable treatments for cancer, but its clinical application is limited due to adverse side effects. Melatonin is a potent antioxidant and antiageing molecule, is nontoxic, and enhances the efficacy and reduces the side effects of chemotherapy. In this review, we first summarize the mitochondrial protective role of melatonin in the context of chemotherapeutic drug-induced toxicity. Thereafter, we tabulate the protective actions of melatonin against ageing and the harmful roles induced by chemotherapy and chemotherapeutic agents, including anthracyclines, alkylating agents, platinum, antimetabolites, mitotic inhibitors, and molecular-targeted agents. Finally, we discuss several novel directions for future research in this area. The information compiled in this review will provide a comprehensive reference for the protective activities of melatonin in the context of chemotherapy drug-induced toxicity and will contribute to the design of future studies and increase the potential of melatonin as a therapeutic agent.
Collapse
|
8
|
Detection of Increased Relative Expression Units of Bacteroides and Prevotella, and Decreased Clostridium leptum in Stool Samples from Brazilian Rheumatoid Arthritis Patients: A Pilot Study. Microorganisms 2019; 7:microorganisms7100413. [PMID: 31581593 PMCID: PMC6843655 DOI: 10.3390/microorganisms7100413] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 09/25/2019] [Indexed: 12/27/2022] Open
Abstract
Interactions between gut microbes and disease modifying antirheumatic drugs (DMARDs) have been proposed. The aim of the present study was to evaluate the presence of some specific bacteria in stool samples from Brazilian RA patients receiving DMARDs and correlate these data with diet, clinical parameters, and cytokines. Stool samples were used for gut bacteria evalutation by qPCR. Serum samples were used to quantify IL-4 and IL-10 by flow cytometer. Statistics were performed by Pearson chi-square, Mann–Whitney U test, and Spearman’s correlation. The study included 20 RA patients and 30 healthy controls. There were no significant differences (p > 0.05) in dietary habits between RA patients and controls. Concerning gut bacteria, we observed an increase in relative expression units (REU) of Bacteroides and Prevotella species in stool samples from patients, and a decrease in REU of Clostridium leptum when compared with healthy controls. Positive correlation between Prevotella and rheumatoid factor was detected. The IL-4 and IL-10 concentrations were increased in patients when compared with controls. We concluded that gut bacteria are different between RA patients receiving DMARDs and healthy controls. Further studies are necessary to determine the real role of gut microbes and their metabolities in clinical response to different DMARDs in RA patients.
Collapse
|
9
|
Nitric oxide plays a critical role in methotrexate-induced hyperplasia of enterochromaffin cells containing 5-hydroxytryptamine in rat small intestine. J Pharmacol Sci 2019; 141:32-40. [DOI: 10.1016/j.jphs.2019.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 08/18/2019] [Accepted: 09/03/2019] [Indexed: 12/24/2022] Open
|
10
|
Li S, Wang Y, Chen L, Wang Z, Liu G, Zuo B, Liu C, Sun D. Beraprost sodium mitigates renal interstitial fibrosis through repairing renal microvessels. J Mol Med (Berl) 2019; 97:777-791. [PMID: 30923844 DOI: 10.1007/s00109-019-01769-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 02/12/2019] [Accepted: 03/05/2019] [Indexed: 12/17/2022]
Abstract
Beraprost sodium (BPS), as a prostacyclin analog, plays a significant role in various diseases based on its antiplatelet and vasodilation functions. However, its regulation and role in chronic kidney disease (CKD) still remain elusive. Here, we determined whether BPS could alleviate renal interstitial fibrosis, and improve the renal function and its therapeutic mechanism. In vitro, BPS increased angiogenesis in the HUVECs incubated with BPS detected by tube formation assay and repair damaged endothelial cell-cell junctions induced by hypoxia. In vivo, mice were randomly assigned to a sham-operation group (sham), a unilateral ureteral obstruction group (UUO), and a BPS intragastrical administration group (BPS), and sacrificed at days 3 and 7 post-surgery (six in each group). In UUO model, tissue hypoxia, renal inflammation, oxidative stress, and fibrotic lesions were detected by q-PCR and Western blot techniques and peritubular capillaries (PTCs) injury was detected by a novel technique of fluorescent microangiography (FMA) and analyzed by MATLAB software. Meanwhile, we identified cells undergoing endothelial cell-to-myofibroblast transition by the coexpression of endothelial cell (CD31) and myofibroblast (a-SMA) markers in the obstructed kidney. In contrast, BPS protected against interstitial fibrosis and substantially reduced the number of endothelial cell-to-myofibroblast transition cells. In conclusion, our data indicate the potent therapeutic of BPS in mitigating fibrosis through repairing renal microvessels and suppressing endothelial-mesenchymal transition (EndMT) progression after inhibiting inflammatory and oxidative stress effects. KEY MESSAGES: BPS could improve renal recovery through anti-inflammatory and anti-oxidative pathways. BPS could mitigate fibrosis through repairing renal microvessels and suppressing endothelial-mesenchymal transition (EndMT).
Collapse
Affiliation(s)
- Shulin Li
- Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, 221002, China.,Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China
| | - Yanping Wang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China
| | - Lu Chen
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China
| | - Zhuojun Wang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China
| | - Guodong Liu
- Department of Orthopedics, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221006, China
| | - Bangjie Zuo
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China
| | - Caixia Liu
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China.
| | - Dong Sun
- Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, 221002, China. .,Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China.
| |
Collapse
|
11
|
Li S, Zhao Y, Wang Z, Wang J, Liu C, Sun D. Transplantation of Amniotic Fluid-Derived Stem Cells Preconditioned with Glial Cell Line-Derived Neurotrophic Factor Gene Alleviates Renal Fibrosis. Cell Transplant 2018; 28:65-78. [PMID: 30497277 PMCID: PMC6322139 DOI: 10.1177/0963689718815850] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Amniotic fluid-derived stem cells (AFSCs), which exhibit both embryonic and mesenchymal stem cell characteristics, have been shown to mitigate the degree of renal interstitial fibrosis. The aim of the present study was to determine whether transplantation of glial cell line-derived neurotrophic factor (GDNF)–modified AFSCs is more useful than transplantation of unmodified AFSCs for the treatment of renal interstitial fibrosis. Mice were randomly assigned to a sham-operation group (sham), a unilateral ureteral obstruction (UUO)-saline solution group (UUO), an AFSC transplantation group (AFSC) and a GDNF-modified AFSC transplantation group (GDNF-AFSC) and sacrificed at days 3 and 7 post-surgery (six in each group). We showed that GDNF-AFSCs noticeably suppressed oxidative stress and inflammation; additionally, GDNF-AFSCs positively regulated peritubular capillaries (PTCs), vascular endothelial growth factor (VEGF), hypoxia inducible factor-1α (HIF-1α), and transforming growth factor-β1 (TGF-β1) protein levels. Transmission electron microscopy (TEM) revealed that mitochondrial injury induced by the UUO model was significantly ameliorated after the mice were treated with GDNF-AFSCs. Therefore, we determined that GDNF gene promotes the abilities of AFSCs to inhibit inflammatory and oxidative stress effects, repair renal microvessels, relieve tissue hypoxia and mitochondrial damage, and, ultimately, alleviate renal interstitial fibrosis.
Collapse
Affiliation(s)
- Shulin Li
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yuan Zhao
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhuojun Wang
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Jia Wang
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Caixia Liu
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Dong Sun
- 1 Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,2 Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
12
|
Natarajan K, Abraham P, Kota R, Isaac B. NF-κB-iNOS-COX2-TNF α inflammatory signaling pathway plays an important role in methotrexate induced small intestinal injury in rats. Food Chem Toxicol 2018; 118:766-783. [DOI: 10.1016/j.fct.2018.06.040] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Revised: 06/16/2018] [Accepted: 06/19/2018] [Indexed: 12/21/2022]
|
13
|
Abozaid OAR, Moawed FSM, Farrag MA, Abdel Aziz AAA. 4-(4-Hydroxy-3-methoxyphenyl)-2-butanone modulates redox signal in gamma-irradiation-induced nephrotoxicity in rats. Free Radic Res 2017; 51:943-953. [DOI: 10.1080/10715762.2017.1395025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Omayma A. R. Abozaid
- Department of Biochemistry, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| | - Fatma S. M. Moawed
- Health Radiation Research, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | - Mostafa A. Farrag
- Radiation Biology, National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | | |
Collapse
|
14
|
Maladaptive DNA repair is the ultimate contributor to the death of trimethoprim-treated cells under aerobic and anaerobic conditions. Proc Natl Acad Sci U S A 2017; 114:11512-11517. [PMID: 29073080 PMCID: PMC5664507 DOI: 10.1073/pnas.1706236114] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Antibiotic resistance leads to substantial mortality and morbidity and significant economic cost because it seriously undermines our ability to treat bacterial infections. Therefore, a better understanding of the effect of antibiotics on bacteria is needed to increase the effectiveness of treatments and slow the emergence of resistance. The bactericidal effects of antibiotics are triggered by target-specific interactions, but there is growing evidence that an important part of their cytotoxicity results from metabolic disturbances induced by treatment. In this article, we report that the perturbation of DNA replication by a wide-spectrum antibiotic, trimethoprim, affects bacterial metabolism, which provokes the production of genotoxic agents and DNA damage, whose processing ultimately contributes to cell death under both aerobic and anaerobic conditions. The bactericidal effects of antibiotics are undoubtedly triggered by target-specific interactions, but there is growing evidence that an important aspect of cytotoxicity results from treatment-induced metabolic perturbations. In this study, we characterized molecular mechanisms whereby trimethoprim treatment results in cell death, using Escherichia coli as the model organism. E. coli cells grown in rich medium that contained all amino acids and low amounts of thymidine were treated with trimethoprim under aerobic and anaerobic conditions. Under these growth conditions, accelerated thymine depletion is the primary trigger of the processes leading to cell death. Thymine depletion-induced DNA replication stress leads to the production of reactive oxygen species under aerobic conditions and of the DNA-damaging byproducts of nitrate respiration under anaerobic conditions. Lowering the DNA replication initiation rate by introducing the dnaA(Sx) allele or by overexpressing Hda protein reduces the number of active replication forks, which reduces the consumption of thymidine and increases resistance to trimethoprim under both aerobic and anaerobic conditions. Analysis of the involvement of DNA repair enzymes in trimethoprim-induced cytotoxicity clearly indicates that different amounts and/or different types of DNA lesions are produced in the presence or absence of oxygen. Maladaptive processing of the DNA damage by DNA repair enzymes, in particular by MutM and MutY DNA glycosylases, ultimately contributes to cell death.
Collapse
|
15
|
Natarajan K, Abraham P, Kota R. Activation of the mitochondrial apoptotic pathway contributes to methotrexate-induced small intestinal injury in rats. Cell Biochem Funct 2017; 35:378-391. [DOI: 10.1002/cbf.3285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 04/12/2017] [Accepted: 07/07/2017] [Indexed: 01/05/2023]
Affiliation(s)
- Kasthuri Natarajan
- Department of Biochemistry; Christian Medical College Campus; Vellore Tamil Nadu India
| | - Premila Abraham
- Department of Biochemistry; Christian Medical College Campus; Vellore Tamil Nadu India
| | - Rekha Kota
- Department of Pathology; Madha Medical College Thandalam; Chennai Tamil Nadu India
| |
Collapse
|
16
|
Natarajan K, Abraham P, Kota R, Selvakumar D. Aminoguanidine pretreatment prevents methotrexate-induced small intestinal injury in the rat by attenuating nitrosative stress and restoring the activities of vital mitochondrial enzymes. J Basic Clin Physiol Pharmacol 2017; 28:239-247. [PMID: 28099126 DOI: 10.1515/jbcpp-2016-0099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 10/26/2016] [Indexed: 06/06/2023]
Abstract
BACKGROUND One of the major toxic side effects of methotrexate (MTX) is enterocolitis, for which there is no efficient standard treatment. Nitric oxide overproduction has been reported to play an important role in MTX-induced mucositis. This study was designed to investigate whether pretreatment with aminoguanidine (AG) - a selective iNOS inhibitor - prevents MTX-induced mucositis in rats. METHODS Rats were pretreated with AG (30 and 50 mg/kg body weight) i.p. daily 1 h before MTX (7 mg/kg body weight) administration for 3 consecutive days. After the final dose of MTX, the rats were killed, and the small intestines were used for analysis. RESULTS The small intestines of MTX-treated rats showed moderate to severe injury. Pretreatment with AG had a dose-dependent protective effect on MTX-induced mucositis. AG pretreatment reduced iNOS protein levels, mucosal nitric oxide levels, and protein tyrosine nitration. AG pretreatment also restored the activities of electron transport chain (ETC) complexes, vital tricarboxylic acid (TCA cycle) enzymes, and mitochondrial antioxidant enzymes. CONCLUSIONS These findings suggest that AG is beneficial in ameliorating MTX-induced enteritis in rats.
Collapse
Affiliation(s)
- Kasthuri Natarajan
- Department of Biochemistry, Christian Medical College, Bagayam, Vellore, Tamil Nadu
| | - Premila Abraham
- Department of Biochemistry, Christian Medical College, Bagayam, Vellore 632002, Tamil Nadu
| | - Rekha Kota
- Department of Pathology, Madha Medical College, Thandalam, Kovur, Chennai, Tamil Nadu
| | - Dhayakani Selvakumar
- Department of Biochemistry, Christian Medical College, Bagayam, Vellore, Tamil Nadu
| |
Collapse
|
17
|
Nardini C, Devescovi V, Liu Y, Zhou X, Lu Y, Dent JE. Systemic Wound Healing Associated with local sub-Cutaneous Mechanical Stimulation. Sci Rep 2016; 6:39043. [PMID: 28008941 PMCID: PMC5180236 DOI: 10.1038/srep39043] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 11/17/2016] [Indexed: 12/12/2022] Open
Abstract
Degeneration is a hallmark of autoimmune diseases, whose incidence grows worldwide. Current therapies attempt to control the immune response to limit degeneration, commonly promoting immunodepression. Differently, mechanical stimulation is known to trigger healing (regeneration) and it has recently been proposed locally for its therapeutic potential on severely injured areas. As the early stages of healing consist of altered intra- and inter-cellular fluxes of soluble molecules, we explored the potential of this early signal to spread, over time, beyond the stimulation district and become systemic, to impact on distributed or otherwise unreachable injured areas. We report in a model of arthritis in rats how stimulations delivered in the subcutaneous dorsal tissue result, over time, in the control and healing of the degeneration of the paws' joints, concomitantly with the systemic activation of wound healing phenomena in blood and in correlation with a more eubiotic microbiome in the gut intestinal district.
Collapse
Affiliation(s)
- Christine Nardini
- Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai 200031, P.R. China
- CNR IAC “Mauro Picone”, Via dei Taurini 19 00185-Roma, Italy
| | - Valentina Devescovi
- Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai 200031, P.R. China
| | - Yuanhua Liu
- Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai 200031, P.R. China
- Bioinformatics Platform, Institut Pasteur of Shanghai, CAS, Shanghai 200031, P.R. China
| | - Xiaoyuan Zhou
- Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai 200031, P.R. China
| | - Youtao Lu
- Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai 200031, P.R. China
| | - Jennifer E. Dent
- Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai 200031, P.R. China
- NORSAS consultancy limited, Norwich (NR12 8QP), Norfolk, UK
| |
Collapse
|
18
|
Zhou X, Nardini C. A method for automated pathogenic content estimation with application to rheumatoid arthritis. BMC SYSTEMS BIOLOGY 2016; 10:107. [PMID: 27846901 PMCID: PMC5111251 DOI: 10.1186/s12918-016-0344-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 10/13/2016] [Indexed: 11/29/2022]
Abstract
Background Sequencing technologies applied to mammals’ microbiomes have revolutionized our understanding of health and disease. Hence, to assess diseases’ progression as well as therapies longterm effects, the impact of maladies and drugs on the gut-intestinal (GI) microbiome has to be evaluated. Typical metagenomic analyses are run to associate to a condition (disease, therapy, diet) a pool of bacteria, whose eubiotic/dysbiotic potential is assessed either by α-diversity, a measure of the varieties populating the microbiome, or by Firmicutes to Bacteroides ratio, associated to systemic inflammation, and finally by manual and direct inspection of bacteria’s biological functions, when known. These approaches lead to results sometimes difficult to interpret in terms of the evolution towards a specific microbial composition, harmed by large areas of unknown. Results We propose to additionally evaluate a microbiome based on its global composition, by automatic annotation of pathogenic genera and statistical assessment of the net varied frequency of harmless versus harmful organisms. This application is intuitive, quantitative and computationally efficient and designed to cope with the currently incomplete species’ functional knowledge. Our results, applied to human GI-microbiome data exemplify how this layer of information provides additional insights into treatments’ impact on the GI microbiome, allowing to characterize a more physiologic effects of Prednisone versus Methotrexate, two treatments for rheumatoid arthritis (RA) a complex autoimmune systemic disease. Conclusions Our quantitative analysis integrates with previous approaches offering an additional systemic level of interpretation here applied, for its potential to translate into clinically relevant information, to the therapies for RA. Electronic supplementary material The online version of this article (doi:10.1186/s12918-016-0344-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoyuan Zhou
- Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Christine Nardini
- Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Shanghai, People's Republic of China. .,University of Chinese Academy of Sciences, Beijing, People's Republic of China. .,Personalgenomics, Verona, Italy.
| |
Collapse
|
19
|
Shi CJ, Wen XS, Gao HF, Liu ZH, Xu XK, Li LF, Shen T, Xian CJ. Steamed root of Rehmannia glutinosa Libosch (Plantaginaceae) alleviates methotrexate-induced intestinal mucositis in rats. JOURNAL OF ETHNOPHARMACOLOGY 2016; 183:143-150. [PMID: 26934449 DOI: 10.1016/j.jep.2016.02.035] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 01/20/2016] [Accepted: 02/25/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Intestinal mucositis induced by chemotherapy is a severe clinical problem in cancer patients that currently lack effective interventions. In traditional Chinese medicine, chemotherapeutic toxicity is diagnosed as Qi and Yin deficiency, and steamed rehmannia root (SRR) is frequently prescribed to these patients. Whether SRR can prevent the adverse effects remains to be confirmed experimentally. The present study used a rat model to investigate potential efficacy and action mechanisms of SRR in attenuating the adverse effects caused by chemotherapy. MATERIALS AND METHODS Intraperitoneal injection of a single dose of anti-metabolite methotrexate (MTX, 25mg/kg) was given to adult Wistar rats, which also received oral gavage of water or SRR (1.08g/kg twice daily 3 days before and 4 days after MTX treatment), or calcium folinate (CF, a clinically used MTX antidote as a comparison, at 1mg/kg twice daily 36h after MTX treatment), or SRR and CF in combination. Animals were sacrificed 4 days after MTX treatment. Complete blood cell counting was carried out. Jejunum was analyzed histologically for mucosal damage, immunohistochemically for proliferating cell nuclear antigen (PCNA), and biochemically for thiobarbituric acid reactive substances (TBARS) and reduced glutathione (GSH), as well as for tumor necrosis factor alpha (TNF-α). RESULTS MTX treatment led to weight loss, leucopenia, polycythemia, increase in large thrombocyte ratio, intestinal villus atrophy, crypt loss and reduction in PCNA positive crypt cells, increases in mucosal TBARS and TNF-α and decrease in GSH. All these alterations were inhibited by SRR administration except leucopenia, and the effects of CF or CF plus SRR supplementation were found to be inferior to those of SRR. CONCLUSIONS SRR can alleviate MTX-induced gut mucositis, which may be achieved by inhibiting MTX-induced oxidative stress and inflammatory response. These findings support the application of SRR in chemotherapy but not the combined application of SRR and CF.
Collapse
Affiliation(s)
- Cheng-Jin Shi
- Institute of Pharmacognosy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xue-Sen Wen
- Institute of Pharmacognosy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China.
| | - Hui-Feng Gao
- Institute of Pharmacognosy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Zhi-Hua Liu
- Institute of Pharmacognosy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xu-Kang Xu
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia
| | - Li-Fen Li
- Institute of Pharmacognosy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Tao Shen
- Institute of Pharmacognosy, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Cory J Xian
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA 5001, Australia
| |
Collapse
|
20
|
Natarajan K, Abraham P. Methotrexate administration induces differential and selective protein tyrosine nitration and cysteine nitrosylation in the subcellular organelles of the small intestinal mucosa of rats. Chem Biol Interact 2016; 251:45-59. [DOI: 10.1016/j.cbi.2016.03.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/11/2016] [Accepted: 03/29/2016] [Indexed: 12/26/2022]
|
21
|
Antioxidant defence systems in the protozoan pathogen Giardia intestinalis. Mol Biochem Parasitol 2016; 206:56-66. [DOI: 10.1016/j.molbiopara.2015.12.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/26/2015] [Accepted: 12/01/2015] [Indexed: 01/03/2023]
|
22
|
Pioglitazone ameliorates methotrexate-induced renal endothelial dysfunction via amending detrimental changes in some antioxidant parameters, systemic cytokines and Fas production. Vascul Pharmacol 2015; 74:139-150. [DOI: 10.1016/j.vph.2015.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Revised: 06/29/2015] [Accepted: 07/04/2015] [Indexed: 02/07/2023]
|
23
|
Vanhoecke B, Bateman E, Mayo B, Vanlancker E, Stringer A, Thorpe D, Keefe D. Dark Agouti rat model of chemotherapy-induced mucositis: establishment and current state of the art. Exp Biol Med (Maywood) 2015; 240:725-41. [PMID: 25966981 PMCID: PMC4935219 DOI: 10.1177/1535370215581309] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mucositis is a major oncological problem. The entire gastrointestinal and genitourinary tract and also other mucosal surfaces can be affected in recipients of radiotherapy, and/or chemotherapy. Major progress has been made in recent years in understanding the mechanisms of oral and small intestinal mucositis, which appears to be more prominent than colonic damage. This progress is largely due to the development of representative laboratory animal models of mucositis. This review focuses on the development and establishment of the Dark Agouti rat mammary adenocarcinoma model by the Mucositis Research Group of the University of Adelaide over the past 20 years to characterize the mechanisms underlying methotrexate-, 5-fluorouracil-, and irinotecan-induced mucositis. It also aims to summarize the results from studies using different animal model systems to identify new molecular and cellular markers of mucositis.
Collapse
Affiliation(s)
- Barbara Vanhoecke
- Mucositis Research Group, Centre for Personalised Cancer Medicine (CPCM), Centre for Clinical Research Excellence (CCRE) in Oral Health, Faculty of Health Sciences, University of Adelaide, Adelaide, 5005 South Australia, Australia Laboratory of Microbial Ecology and Technology, University of Ghent, 9000 Ghent, Belgium
| | - Emma Bateman
- Mucositis Research Group, Centre for Personalised Cancer Medicine (CPCM), Centre for Clinical Research Excellence (CCRE) in Oral Health, Faculty of Health Sciences, University of Adelaide, Adelaide, 5005 South Australia, Australia
| | - Bronwen Mayo
- Mucositis Research Group, Centre for Personalised Cancer Medicine (CPCM), Centre for Clinical Research Excellence (CCRE) in Oral Health, Faculty of Health Sciences, University of Adelaide, Adelaide, 5005 South Australia, Australia Sansom Institute for Health Research, University of South Australia, Adelaide, 5001 South Australia, Australia
| | - Eline Vanlancker
- Laboratory of Microbial Ecology and Technology, University of Ghent, 9000 Ghent, Belgium
| | - Andrea Stringer
- Sansom Institute for Health Research, University of South Australia, Adelaide, 5001 South Australia, Australia
| | - Daniel Thorpe
- Sansom Institute for Health Research, University of South Australia, Adelaide, 5001 South Australia, Australia
| | - Dorothy Keefe
- Mucositis Research Group, Centre for Personalised Cancer Medicine (CPCM), Centre for Clinical Research Excellence (CCRE) in Oral Health, Faculty of Health Sciences, University of Adelaide, Adelaide, 5005 South Australia, Australia Director, SA Cancer Service, Royal Adelaide Hospital, Adelaide, 5005 South Australia, Australia
| |
Collapse
|
24
|
Tieri P, Zhou X, Zhu L, Nardini C. Multi-omic landscape of rheumatoid arthritis: re-evaluation of drug adverse effects. Front Cell Dev Biol 2014; 2:59. [PMID: 25414848 PMCID: PMC4220167 DOI: 10.3389/fcell.2014.00059] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/26/2014] [Indexed: 12/19/2022] Open
Abstract
Objective: To provide a frame to estimate the systemic impact (side/adverse events) of (novel) therapeutic targets by taking into consideration drugs potential on the numerous districts involved in rheumatoid arthritis (RA) from the inflammatory and immune response to the gut-intestinal (GI) microbiome. Methods: We curated the collection of molecules from high-throughput screens of diverse (multi-omic) biochemical origin, experimentally associated to RA. Starting from such collection we generated RA-related protein-protein interaction (PPI) networks (interactomes) based on experimental PPI data. Pharmacological treatment simulation, topological and functional analyses were further run to gain insight into the proteins most affected by therapy and by multi-omic modeling. Results: Simulation on the administration of MTX results in the activation of expected (apoptosis) and adverse (nitrogenous metabolism alteration) effects. Growth factor receptor-bound protein 2 (GRB2) and Interleukin-1 Receptor Associated Kinase-4 (IRAK4, already an RA target) emerge as relevant nodes. The former controls the activation of inflammatory, proliferative and degenerative pathways in host and pathogens. The latter controls immune alterations and blocks innate response to pathogens. Conclusions: This multi-omic map properly recollects in a single analytical picture known, yet complex, information like the adverse/side effects of MTX, and provides a reliable platform for in silico hypothesis testing or recommendation on novel therapies. These results can support the development of RA translational research in the design of validation experiments and clinical trials, as such we identify GRB2 as a robust potential new target for RA for its ability to control both synovial degeneracy and dysbiosis, and, conversely, warn on the usage of IRAK4-inhibitors recently promoted, as this involves potential adverse effects in the form of impaired innate response to pathogens.
Collapse
Affiliation(s)
- Paolo Tieri
- IAC - Istituto per le Applicazioni del Calcolo "Mauro Picone," CNR - Consiglio Nazionale delle Ricerche Rome, Italy ; Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, Chinese Academy of Sciences - Max Planck Society Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences Shanghai, China
| | - XiaoYuan Zhou
- Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, Chinese Academy of Sciences - Max Planck Society Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences Shanghai, China
| | - Lisha Zhu
- Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, Chinese Academy of Sciences - Max Planck Society Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences Shanghai, China
| | - Christine Nardini
- Group of Clinical Genomic Networks, Key Laboratory of Computational Biology, Chinese Academy of Sciences - Max Planck Society Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences Shanghai, China
| |
Collapse
|
25
|
Takano Y, Machida T, Obara Y, Hirano M, Kudo S, Takagi M, Hamaue N, Iizuka K, Hirafuji M. Methotrexate causes a change in intestinal 5-hydroxytryptamine metabolism in rats. Eur J Pharmacol 2014; 740:496-503. [DOI: 10.1016/j.ejphar.2014.06.038] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/19/2014] [Accepted: 06/19/2014] [Indexed: 11/15/2022]
|
26
|
Mastronicola D, Falabella M, Testa F, Pucillo LP, Teixeira M, Sarti P, Saraiva LM, Giuffrè A. Functional characterization of peroxiredoxins from the human protozoan parasite Giardia intestinalis. PLoS Negl Trop Dis 2014; 8:e2631. [PMID: 24416465 PMCID: PMC3886907 DOI: 10.1371/journal.pntd.0002631] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 11/26/2013] [Indexed: 01/03/2023] Open
Abstract
The microaerophilic protozoan parasite Giardia intestinalis, causative of one of the most common human intestinal diseases worldwide, infects the mucosa of the proximal small intestine, where it has to cope with O2 and nitric oxide (NO). Elucidating the antioxidant defense system of this pathogen lacking catalase and other conventional antioxidant enzymes is thus important to unveil novel potential drug targets. Enzymes metabolizing O2, NO and superoxide anion (O2−•) have been recently reported for Giardia, but it is yet unknown how the parasite copes with H2O2 and peroxynitrite (ONOO−). Giardia encodes two yet uncharacterized 2-cys peroxiredoxins (Prxs), GiPrx1a and GiPrx1b. Peroxiredoxins are peroxidases implicated in virulence and drug resistance in several parasitic protozoa, able to protect from nitroxidative stress and repair oxidatively damaged molecules. GiPrx1a and a truncated form of GiPrx1b (deltaGiPrx1b) were expressed in Escherichia coli, purified and functionally characterized. Both Prxs effectively metabolize H2O2 and alkyl-hydroperoxides (cumyl- and tert-butyl-hydroperoxide) in the presence of NADPH and E. coli thioredoxin reductase/thioredoxin as the reducing system. Stopped-flow experiments show that both proteins in the reduced state react with ONOO− rapidly (k = 4×105 M−1 s−1 and 2×105 M−1 s−1 at 4°C, for GiPrx1a and deltaGiPrx1b, respectively). Consistent with a protective role against oxidative stress, expression of GiPrx1a (but not deltaGiPrx1b) is induced in parasitic cells exposed to air O2 for 24 h. Based on these results, GiPrx1a and deltaGiPrx1b are suggested to play an important role in the antioxidant defense of Giardia, possibly contributing to pathogenesis. Giardia intestinalis causes one of the most common human intestinal diseases worldwide, called giardiasis. This microorganism infects the small intestine where it has to cope with O2, nitric oxide (NO) and related reactive species that are toxic for Giardia as it lacks most of the conventional antioxidant enzymes. Understanding how this pathogen survives oxidative stress is thus important because it may help to identify novel drug targets to combat giardiasis. Some enzymes playing a role in the antioxidant defense of Giardia have been recently reported, but it is yet unknown how the parasite copes with two well-known oxidants, hydrogen peroxide (H2O2) and peroxynitrite (ONOO−). In this study, the Authors show that Giardia expresses two enzymes (called peroxiredoxins), yet uncharacterized, that are able not only to degrade both H2O2 and ONOO−, but also to repair damaged molecules (called hydroperoxides) that accumulate in the cell under oxidative stress conditions. These results are totally unprecedented because no enzymes with these types of functions have been reported for Giardia to date. If these two enzymes will prove to be essential for Giardia virulence in future studies, a new way will be paved towards the discovery of novel drugs to treat giardiasis.
Collapse
Affiliation(s)
| | - Micol Falabella
- Department of Biochemical Sciences and Istituto Pasteur – Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy
| | - Fabrizio Testa
- Department of Biochemical Sciences and Istituto Pasteur – Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy
| | | | - Miguel Teixeira
- Instituto de Tecnologia Quimica e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Paolo Sarti
- CNR Institute of Molecular Biology and Pathology, Rome, Italy
- Department of Biochemical Sciences and Istituto Pasteur – Fondazione Cenci Bolognetti, Sapienza University of Rome, Italy
| | - Lígia M. Saraiva
- Instituto de Tecnologia Quimica e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | | |
Collapse
|
27
|
Acipayam C, Bayram I, Daglioglu K, Doran F, Yilmaz S, Sezgin G, Totan Ateş B, Ozkan A, Tanyeli A. The protective effect of hesperidin on methotrexate-induced intestinal epithelial damage in rats: an experimental study. Med Princ Pract 2014; 23:45-52. [PMID: 24247210 PMCID: PMC5586843 DOI: 10.1159/000355900] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 09/22/2013] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE The purpose of this experimental study was to evaluate the efficacy of hesperidin (HES) in protecting against methotrexate (MTX)-induced intestinal damage using histopathological and immunohistochemical techniques. MATERIALS AND METHODS Seventy-eight male Wistar albino rats were divided into 4 groups that received (a) saline only (control group), n = 19; (b) HES only, n = 19; (c) MTX only, n = 19, and (d) MTX plus HES, n = 21. On the first day of the study, a single dose of MTX (20 mg/kg) was administered intraperitoneally to group 3 and 4 rats. The HES (200 mg/kg) was administered by gavage for 5 days. For the MTX plus HES group, HES (200 mg/kg) was administered by gavage for 5 days after MTX treatment. Rats were sacrificed on the 2nd, 4th and 6th day of the study. Tissue samples from the jejunum were taken for histopathological and immunohistochemical analysis. RESULTS On the 4th day, crypt injury in the MTX plus HES group (1.00 ± 0.00) was less than that in the MTX group (2.00 ± 0.89; p < 0.05). The small intestinal damage score was lower in the MTX plus HES group (6.33 ± 0.82) as compared to the MTX group (8.00 ± 2.37). Inducible nitric oxide synthase and interleukin-8 levels were lower in the MTX plus HES group (65 and 25%, respectively) as compared to the corresponding values of the MTX group (80 and 52.5%, respectively). On the 6th day, the Ki-67 proliferation index in the MTX group (45%) was lower than that in the MTX plus HES group (76.67%) and the control group (p < 0.05). The small intestinal damage score was high in the HES group on the 4th day due to increased cellular infiltration. On the 6th day, the Ki-67 proliferation index rose in parallel with the decrease in cellular infiltration and therefore histopathological scoring. The proliferation-enhancing effect of HES also appeared in healthy rats. CONCLUSION HES seemed to have a protective effect against MTX-induced intestinal injury.
Collapse
Affiliation(s)
- Can Acipayam
- Department of Pediatric Hematology/Oncology, Adana, Turkey
- *Can Acıpayam, MD, Department of Pediatric Oncology/BMT Unit, Cukurova University School of Medicine, TR-01330 Balcali, Adana (Turkey), E-Mail
| | - Ibrahim Bayram
- Department of Pediatric Hematology/Oncology, Adana, Turkey
| | | | - Figen Doran
- Department of Pathology, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Sema Yilmaz
- Department of Pediatric Hematology/Oncology, Adana, Turkey
| | - Gülay Sezgin
- Department of Pediatric Hematology/Oncology, Adana, Turkey
| | - Berna Totan Ateş
- Department of Pathology, Faculty of Medicine, Cukurova University, Adana, Turkey
| | - Ayşe Ozkan
- Department of Pediatric Hematology/Oncology, Adana, Turkey
| | - Atila Tanyeli
- Department of Pediatric Hematology/Oncology, Adana, Turkey
| |
Collapse
|
28
|
Kolli VK, Natarajan K, Isaac B, Selvakumar D, Abraham P. Mitochondrial dysfunction and respiratory chain defects in a rodent model of methotrexate-induced enteritis. Hum Exp Toxicol 2013; 33:1051-65. [PMID: 24347301 DOI: 10.1177/0960327113515503] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The efficacy of methotrexate (MTX), a widely used chemotherapeutic drug, is limited by its gastrointestinal toxicity and the mechanism of which is not clear. The present study investigates the possible role of mitochondrial damage in MTX-induced enteritis. Small intestinal injury was induced in Wistar rats by the administration of 7 mg kg(-1) body wt. MTX intraperitoneally for 3 consecutive days. MTX administration resulted in severe small intestinal injury and extensive damage to enterocyte mitochondria. Respiratory control ratio, the single most useful and reliable test of mitochondrial function, and 3-(4,5-dimethylthiazol-2-yll)-2,5-diphenyltetrazolium bromide reduction, a measure of cell viability were significantly reduced in all the fractions of MTX-treated rat enterocytes. A massive decrease (nearly 70%) in the activities of complexes II and IV was also observed. The results of the present study suggest that MTX-induced damage to enterocyte mitochondria may play a critical role in enteritis. MTX-induced alteration in mitochondrial structure may cause its dysfunction and decreases the activities of the electron chain complexes. MTX-induced mitochondrial damage can result in reduced adenosine triphosphate synthesis, thereby interfering with nutrient absorption and enterocyte renewal. This derangement may contribute to malabsorption of nutrients, diarrhea, and weight loss seen in patients on MTX chemotherapy.
Collapse
Affiliation(s)
- V K Kolli
- Department of Biochemistry, Christian Medial College, Bagayam, Vellore, Tamil Nadu, India
| | - K Natarajan
- Department of Biochemistry, Christian Medial College, Bagayam, Vellore, Tamil Nadu, India
| | - B Isaac
- Department of Anatomy, Christian Medial College, Bagayam, Vellore, Tamil Nadu, India
| | - D Selvakumar
- Department of Biochemistry, Christian Medial College, Bagayam, Vellore, Tamil Nadu, India
| | - P Abraham
- Department of Biochemistry, Christian Medial College, Bagayam, Vellore, Tamil Nadu, India
| |
Collapse
|
29
|
A preclinical study on the protective effect of melatonin against methotrexate-induced small intestinal damage: effect mediated by attenuation of nitrosative stress, protein tyrosine nitration, and PARP activation. Cancer Chemother Pharmacol 2013; 71:1209-18. [PMID: 23420439 DOI: 10.1007/s00280-013-2115-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 02/05/2013] [Indexed: 10/27/2022]
Abstract
PURPOSE One of the major toxic side effects of methotrexate (MTX) is enterocolitis. To date, there is no efficient standard treatment for this side effect. Nitrosative stress is reported to play a critical role in MTX-induced mucositis. The purpose of this study is to investigate whether pretreatment with melatonin, an inhibitor of nitro-oxidative stress, prevents MTX-induced mucositis in rats. METHODS Rats were pretreated with melatonin (20 and 40 mg/kg body weight) i.p. daily 1 h before MTX (7 mg/kg body weight) administration for three consecutive days. After the final dose of MTX, the rats were killed and the small intestines were used for analysis. RESULTS The small intestines of MTX-treated rats showed moderate to severe injury. The villi were distorted, blunted, and atrophied and focally absent in various segments of the small intestines. Crypt abscesses were also found, suggesting an inflammatory response. Pretreatment with melatonin had a dose-dependent protective effect on MTX-induced mucositis. Morphology was saved to a moderate extent with 20 mg melatonin pretreatment, and near-normal morphology was achieved with 40 mg melatonin pretreatment. Damage to the villi and crypt abscess was reduced. The villi/crypt ratio was almost restored. Melatonin pretreatment protected the small intestines from MTX-induced damage by attenuating nitrosative stress, protein tyrosine nitration and PARP expression. CONCLUSION Because of its versatility in protecting against nitro-oxidative stress and reducing inflammation, we suggest that melatonin could be beneficial in ameliorating MTX-induced enteritis in humans.
Collapse
|
30
|
Omar NM, Gawish SAAE, Nosseir DA, Sarhan NMR. Protective Effect of Omega-3 Fatty Acids on 5-fluorouracil-induced Small Intestinal Damage in Rats: Histological and Histomorphometric Study. ACTA ACUST UNITED AC 2013. [DOI: 10.3923/tmr.2013.36.62] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
31
|
Gawish SAE, Nosseir D, Omar N, Sarhan N. Histological and Ultra Structural Study of 5-fluorouracil-induced Small Intestinal Mucosal Damage in Rats. ACTA ACUST UNITED AC 2012. [DOI: 10.3923/ajcb.2013.1.21] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
32
|
Kose E, Sapmaz HI, Sarihan E, Vardi N, Turkoz Y, Ekinci N. Beneficial effects of montelukast against methotrexate-induced liver toxicity: a biochemical and histological study. ScientificWorldJournal 2012; 2012:987508. [PMID: 22566785 PMCID: PMC3329899 DOI: 10.1100/2012/987508] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 01/12/2012] [Indexed: 01/16/2023] Open
Abstract
The effects of montelukast against methotrexate-induced liver damage were investigated. 35 Wistar albino female rats were divided into 5 groups as follows: group I: control; group II: montelukast (ML); group III: methotrexate (Mtx); group IV: montelukast treatment after methotrexate application (Mtx + ML); group V: montelukast treatment before methotrexate application (ML + Mtx). At the end of the experiment, the liver tissues of rats were removed. Malondialdehyde (MDA), myeloperoxidase (MPO), and reduced glutathione levels were determined from liver tissues. In addition, the liver tissues were examined histologically. MDA and MPO levels of Mtx group were significantly increased when compared to control group. In Mtx + ML group, these parameters were decreased as compared to Mtx group. Mtx injection exhibited major histological alterations such as eosinophilic staining and swelling of hepatocytes. The glycogen storage in hepatocytes was observed as decreased by periodic acid schiff staining in Mtx group as compared to controls. ML treatment did not completely ameliorate the lesions and milder degenerative alterations as loss of the glycogen content was still present. It was showed that montelukast treatment after methotrexate application could reduce methotrexate-induced experimental liver damage.
Collapse
Affiliation(s)
- Evren Kose
- Department of Anatomy, School of Medicine, İnonu University Medical Faculty, 44280 Malatya, Turkey.
| | | | | | | | | | | |
Collapse
|
33
|
Leitão RF, Brito GA, Oriá RB, Braga-Neto MB, Bellaguarda EA, Silva JV, Gomes AS, Lima-Júnior RC, Siqueira FJ, Freire RS, Vale ML, Ribeiro RA. Role of inducible nitric oxide synthase pathway on methotrexate-induced intestinal mucositis in rodents. BMC Gastroenterol 2011; 11:90. [PMID: 21846355 PMCID: PMC3170268 DOI: 10.1186/1471-230x-11-90] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2011] [Accepted: 08/16/2011] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Methotrexate treatment has been associated to intestinal epithelial damage. Studies have suggested an important role of nitric oxide in such injury. The aim of this study was to investigate the role of nitric oxide (NO), specifically iNOS on the pathogenesis of methotrexate (MTX)-induced intestinal mucositis. METHODS Intestinal mucositis was carried out by three subcutaneous MTX injections (2.5 mg/kg) in Wistar rats and in inducible nitric oxide synthase knock-out (iNOS-/-) and wild-type (iNOS+/+) mice. Rats were treated intraperitoneally with the NOS inhibitors aminoguanidine (AG; 10 mg/Kg) or L-NAME (20 mg/Kg), one hour before MTX injection and daily until sacrifice, on the fifth day. The jejunum was harvested to investigate the expression of Ki67, iNOS and nitrotyrosine by immunohistochemistry and cell death by TUNEL. The neutrophil activity by myeloperoxidase (MPO) assay was performed in the three small intestine segments. RESULTS AG and L-NAME significantly reduced villus and crypt damages, inflammatory alterations, cell death, MPO activity, and nitrotyrosine immunostaining due to MTX challenge. The treatment with AG, but not L-NAME, prevented the inhibitory effect of MTX on cell proliferation. MTX induced increased expression of iNOS detected by immunohistochemistry. MTX did not cause significant inflammation in the iNOS-/- mice. CONCLUSION These results suggest an important role of NO, via activation of iNOS, in the pathogenesis of intestinal mucositis.
Collapse
Affiliation(s)
- Renata Fc Leitão
- Department of Morphology, Federal University of Ceará, Fortaleza, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Yokooji T, Mori N, Murakami T. Modulated pharmacokinetics and increased small intestinal toxicity of methotrexate in bilirubin-treated rats. J Pharm Pharmacol 2011; 63:206-13. [PMID: 21235584 DOI: 10.1111/j.2042-7158.2010.01213.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES The effect of bilirubin treatment on the pharmacokinetics and small intestinal toxicity of methotrexate was evaluated in rats, since bilirubin and its glucuronide conjugates can suppress multidrug resistance-associated protein-mediated transport. METHODS Rats were treated intravenously with bilirubin and the various clearances and tissue distribution of methotrexate were estimated under a steady-state plasma concentration. Intestinal toxicity induced by methotrexate was also evaluated by measuring the leakage of alkaline phosphatase (ALP) activity. Probenecid, an inhibitor for multidrug resistance-associated protein and organic anion transporters, was used for comparison. KEY FINDINGS The treatment with bilirubin increased the steady-state plasma concentration and reduced biliary excretion clearance, urinary excretion clearance and intestinal exsorption clearance of methotrexate, as did treatment with probenecid. The intestinal absorption and jejunum distribution of methotrexate also significantly increased in bilirubin- and probenecid-treated rats. A greater leakage of ALP activity to the luminal fluid, with a lower ALP activity in the intestinal mucosal membrane after intestinal perfusion of methotrexate, was observed in bilirubin- and probenecid-treated rats. CONCLUSIONS Hyperbilirubinemia, which is involved under various disease states, may increase the small intestinal accumulation and toxicities of methotrexate, since high plasma concentrations of conjugated bilirubin can suppress the function of multidrug resistance-associated proteins, which facilitate the efflux of methotrexate out of cells.
Collapse
Affiliation(s)
- Tomoharu Yokooji
- Laboratory of Biopharmaceutics and Pharmacokinetics, Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hiro-koshingai, Kure, Hiroshima, Japan
| | | | | |
Collapse
|
35
|
Intervention of alpha-lipoic acid ameliorates methotrexate-induced oxidative stress and genotoxicity: A study in rat intestine. Chem Biol Interact 2010; 183:85-97. [PMID: 19900424 DOI: 10.1016/j.cbi.2009.10.020] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 10/28/2009] [Accepted: 10/29/2009] [Indexed: 12/18/2022]
Abstract
Methotrexate (MTX) is an anti-metabolite, widely used in the cancer chemotherapy and rheumatoid arthritis. However, its long-term clinical use is restricted on account of its severe intestinal toxicity. The present study was aimed to investigate the intestinal toxicity of MTX and the possible protective effect of alpha-lipoic acid (LA) on Sprague-Dawley rats. MTX-induced intestinal toxicity was evaluated at the dose of 2.5mg/kg for short-term (5 days treatment) and 1mg/kg for long-term (5 days in a week for four consecutive weeks treatment) study. The possible protective effect of LA was evaluated in both short- as well as long-term study in a dose-dependent manner. MTX treatment induced diarrhoea and mortality in rats, indicating its severe toxicity in the target organ of investigation, i.e., intestine. Further, the intestinal toxicity of MTX was assessed by evaluating different parameters of oxidative stress, DNA damage, cytotoxicity as well as histological changes. Immunostaining for p53 revealed higher genotoxic assault in the intestinal cells due to MTX treatment. Pretreatment of rats with LA led to significant decrease in the oxidative stress, DNA damage, cellular damage, inflammatory changes and apoptosis as determined by malondialdehyde level, glutathione level, comet assay parameters, histological evaluation, immunostaining and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL) assay. In the present investigation, we report that LA pretreatment ameliorates MTX-induced intestinal toxicity in rat as evident from the protection against oxidative stress, decrease in DNA damage and protection of cellular morphology as well as improvement in the stool consistency and animal survival rate.
Collapse
|
36
|
Al-Anazi KA, Eltayeb KI, Bakr M, Al-Mohareb FI. Methotrexate-induced acute leukemia: report of three cases and review of the literature. Clin Med Case Rep 2009; 2:43-9. [PMID: 24179373 PMCID: PMC3785348 DOI: 10.4137/ccrep.s3078] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
For many years, methotrexate has been used in the treatment of certain chronic medical disorders e.g. rheumatoid arthritis and psoriasis as well as a number of malignant disorders e.g. acute lymphoblastic leukemia, certain types of lymphoma and breast carcinoma. Its use has been associated with various systemic toxicities and complications. The association between methotrexate therapy and the development of lymphoma and pseudolymphoma is well established. In patients treated with methotrexate, the development of leukemia has been attributed to either the primary disorder e.g. rheumatoid arthritis or to other drugs used concomitantly e.g. cyclophosphamide. Reported here are two patients with rheumatoid arthritis and one patient with psoriasis treated with low dose methotrexate for variable periods of time. Two of these patients developed acute myeloid leukemia on myelodysplastic syndrome background, while the third patient developed pre-B acute lymphoblastic leukemia that expressed few myeloid markers and had a positive philadelphia chromosome. To our knowledge, these are the first reported cases of methotrexate-induced acute leukemia.
Collapse
Affiliation(s)
- Khalid A Al-Anazi
- Section of Adult Hematology and Hematopoietic Stem Cell Transplant, Oncology Centre, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | | | | | | |
Collapse
|
37
|
Hengstler JG, Godoy P, Stewart JD, Bolt HM. Alcohol-induced liver injury: how a small molecule overwhelms one of the cell types with the best regeneration capacity of the human body. Arch Toxicol 2009; 83:513-4. [PMID: 19458936 DOI: 10.1007/s00204-009-0435-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|