351
|
Natural products: potential treatments for cisplatin-induced nephrotoxicity. Acta Pharmacol Sin 2021; 42:1951-1969. [PMID: 33750909 PMCID: PMC8633358 DOI: 10.1038/s41401-021-00620-9] [Citation(s) in RCA: 199] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/01/2021] [Indexed: 12/13/2022]
Abstract
Cisplatin is a clinically advanced and highly effective anticancer drug used in the treatment of a wide variety of malignancies, such as head and neck, lung, testis, ovary, breast cancer, etc. However, it has only a limited use in clinical practice due to its severe adverse effects, particularly nephrotoxicity; 20%–35% of patients develop acute kidney injury (AKI) after cisplatin administration. The nephrotoxic effect of cisplatin is cumulative and dose dependent and often necessitates dose reduction or withdrawal. Recurrent episodes of AKI result in impaired renal tubular function and acute renal failure, chronic kidney disease, uremia, and hypertensive nephropathy. The pathophysiology of cisplatin-induced AKI involves proximal tubular injury, apoptosis, oxidative stress, inflammation, and vascular injury in the kidneys. At present, there are no effective drugs or methods for cisplatin-induced kidney injury. Recent in vitro and in vivo studies show that numerous natural products (flavonoids, saponins, alkaloids, polysaccharide, phenylpropanoids, etc.) have specific antioxidant, anti-inflammatory, and anti-apoptotic properties that regulate the pathways associated with cisplatin-induced kidney damage. In this review we describe the molecular mechanisms of cisplatin-induced nephrotoxicity and summarize recent findings in the field of natural products that undermine these mechanisms to protect against cisplatin-induced kidney damage and provide potential strategies for AKI treatment.
Collapse
|
352
|
Biochemical and anti-inflammatory effect of Ocimum americanum Linn. extracts on gentamicin- and cisplatin-induced nephrotoxicity in rats. ADVANCES IN TRADITIONAL MEDICINE 2021. [DOI: 10.1007/s13596-020-00501-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
353
|
Black LM, Farrell ER, Barwinska D, Osis G, Zmijewska AA, Traylor AM, Esman SK, Bolisetty S, Whipple G, Kamocka MM, Winfree S, Spangler DR, Khan S, Zarjou A, El-Achkar TM, Agarwal A. VEGFR3 tyrosine kinase inhibition aggravates cisplatin nephrotoxicity. Am J Physiol Renal Physiol 2021; 321:F675-F688. [PMID: 34658261 PMCID: PMC8714977 DOI: 10.1152/ajprenal.00186.2021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/24/2022] Open
Abstract
Expansion of renal lymphatic networks, or lymphangiogenesis (LA), is well recognized during development and is now being implicated in kidney diseases. Although LA is associated with multiple pathological conditions, very little is known about its role in acute kidney injury. The purpose of this study was to evaluate the role of LA in a model of cisplatin-induced nephrotoxicity. LA is predominately regulated by vascular endothelial growth factor (VEGF)-C and VEGF-D, ligands that exert their function through their cognate receptor VEGF receptor 3 (VEGFR3). We demonstrated that use of MAZ51, a selective VEGFR3 inhibitor, caused significantly worse structural and functional kidney damage in cisplatin nephrotoxicity. Apoptotic cell death and inflammation were also increased in MAZ51-treated animals compared with vehicle-treated animals following cisplatin administration. Notably, MAZ51 caused significant upregulation of intrarenal phospho-NF-κB, phospho-JNK, and IL-6. Cisplatin nephrotoxicity is associated with vascular congestion due to endothelial dysfunction. Using three-dimensional tissue cytometry, a novel approach to explore lymphatics in the kidney, we detected significant vascular autofluorescence attributed to erythrocytes in cisplatin alone-treated animals. Interestingly, no such congestion was detected in MAZ51-treated animals. We found increased renal vascular damage in MAZ51-treated animals, whereby MAZ51 caused a modest decrease in the endothelial markers endomucin and von Willebrand factor, with a modest increase in VEGFR2. Our findings identify a protective role for de novo LA in cisplatin nephrotoxicity and provide a rationale for the development of therapeutic approaches targeting LA. Our study also suggests off-target effects of MAZ51 on the vasculature in the setting of cisplatin nephrotoxicity.NEW & NOTEWORTHY Little is known about injury-associated LA in the kidney and its role in the pathophysiology of acute kidney injury (AKI). Observed exacerbation of cisplatin-induced AKI after LA inhibition was accompanied by increased medullary damage and cell death in the kidney. LA inhibition also upregulated compensatory expression of LA regulatory proteins, including JNK and NF-κB. These data support the premise that LA is induced during AKI and lymphatic expansion is a protective mechanism in cisplatin nephrotoxicity.
Collapse
Affiliation(s)
- Laurence M Black
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Elisa R Farrell
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daria Barwinska
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
| | - Gunars Osis
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anna A Zmijewska
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Amie M Traylor
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stephanie K Esman
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Subhashini Bolisetty
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Grace Whipple
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Malgorzata M Kamocka
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
| | - Seth Winfree
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
| | - Daryll R Spangler
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Shehnaz Khan
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
| | - Abolfazl Zarjou
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Tarek M El-Achkar
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
- Indiana Center for Biological Microscopy, Indianapolis, Indiana
- Indianapolis Veterans Affairs Medical Center, Indianapolis, Indiana
| | - Anupam Agarwal
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
- Nephrology Research and Training Center, University of Alabama at Birmingham, Birmingham, Alabama
- Birmingham Veterans Administration Medical Center, Birmingham, Alabama
| |
Collapse
|
354
|
Iqbal MO, Sial AS, Akhtar I, Naeem M, Hazafa A, Ansari RA, Rizvi SAA. The nephroprotective effects of Daucus carota and Eclipta prostrata against cisplatin-induced nephrotoxicity in rats. Bioengineered 2021; 12:12702-12721. [PMID: 34949157 PMCID: PMC8810007 DOI: 10.1080/21655979.2021.2009977] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 11/18/2021] [Accepted: 11/18/2021] [Indexed: 02/07/2023] Open
Abstract
The overuse of cisplatin (>50 mg/m2) is limited to nephrotoxicity, ototoxicity, gastrotoxicity, myelosuppression, and allergic reactions. The objective of this study was to investigate the nephroprotective effects of Daucus carota and Eclipta prostrata extracts on cisplatin-induced nephrotoxicity in Wistar albino rats. The study involved male Wistar albino rats of 8 weeks weighing 220-270 g. A single injection of 5 mg/kg was injected into the rats for nephrotoxicity. Rats were divided into four groups based on dose conentrations. Blood and urine samples of rats were collected on the 0, 7th, 14th, and 21st days for nephrological analysis. The results showed that Cis + DC/Cis + EP (600 mg/kg) significantly (p < 0.001) increased the body weight and reduced the kidney weight of cisplatin-induced nephrotoxicity in rats (p < 0.001) as compared to Cis group. The results showed that 600 mg/kg administration of Cis + DC/Cis +EP successfully (p < 0.005) improved the urine and plasmin creatinine, Na, and K level compared to the Cis group. Histopathological results confirmed that Cis + EP/Cis + DC effectively improved the renal abnormalities. It is concluded that the co-administration of Cis + EP extract showed exceptional nephroprotective effects at a dose rate of 600 mg/kg.
Collapse
Affiliation(s)
- Muhammad Omer Iqbal
- Shandong Provincial Key Laboratory of Glycoscience and Glycoengineering, School of Medicine and Pharmacy, Ocean University of China, Qingdao, Shandong, China
| | - Asad Saleem Sial
- Department of Pharmacognosy, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi, Pakistan
| | - Imran Akhtar
- Department of Pharmacology, Faculty of Pharmacy and Alternative Medicine, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Muhammad Naeem
- College of Life Science, Hebei Normal University, Shijiazhuang, Hebei, China
| | - Abu Hazafa
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Rais A. Ansari
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Syed A. A. Rizvi
- Department of Pharmaceutical Sciences, Hampton University School of Pharmacy, Hampton, VA, USA
| |
Collapse
|
355
|
de Godoy Torso N, Pereira JKN, Visacri MB, Vasconcelos PENS, Loren P, Saavedra K, Saavedra N, Salazar LA, Moriel P. Dysregulated MicroRNAs as Biomarkers or Therapeutic Targets in Cisplatin-Induced Nephrotoxicity: A Systematic Review. Int J Mol Sci 2021; 22:12765. [PMID: 34884570 PMCID: PMC8657822 DOI: 10.3390/ijms222312765] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/07/2021] [Accepted: 10/15/2021] [Indexed: 12/14/2022] Open
Abstract
The purpose of this systematic review was to map out and summarize scientific evidence on dysregulated microRNAs (miRNAs) that can be possible biomarkers or therapeutic targets for cisplatin nephrotoxicity and have already been tested in humans, animals, or cells. In addition, an in silico analysis of the two miRNAs found to be dysregulated in the majority of studies was performed. A literature search was performed using eight databases for studies published up to 4 July 2021. Two independent reviewers selected the studies and extracted the data; disagreements were resolved by a third and fourth reviewers. A total of 1002 records were identified, of which 30 met the eligibility criteria. All studies were published in English and reported between 2010 and 2021. The main findings were as follows: (a) miR-34a and miR-21 were the main miRNAs identified by the studies as possible biomarkers and therapeutic targets of cisplatin nephrotoxicity; (b) the in silico analysis revealed 124 and 131 different strongly validated targets for miR-34a and miR-21, respectively; and (c) studies in humans remain scarce.
Collapse
Affiliation(s)
- Nadine de Godoy Torso
- School of Medical Sciences, University of Campinas, Campinas 13083894, Brazil; (N.d.G.T.); (J.K.N.P.); (M.B.V.); (P.E.N.S.V.)
| | - João Kleber Novais Pereira
- School of Medical Sciences, University of Campinas, Campinas 13083894, Brazil; (N.d.G.T.); (J.K.N.P.); (M.B.V.); (P.E.N.S.V.)
| | - Marília Berlofa Visacri
- School of Medical Sciences, University of Campinas, Campinas 13083894, Brazil; (N.d.G.T.); (J.K.N.P.); (M.B.V.); (P.E.N.S.V.)
| | | | - Pía Loren
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (K.S.); (N.S.); (L.A.S.)
| | - Kathleen Saavedra
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (K.S.); (N.S.); (L.A.S.)
| | - Nicolás Saavedra
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (K.S.); (N.S.); (L.A.S.)
| | - Luis A. Salazar
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (K.S.); (N.S.); (L.A.S.)
| | - Patricia Moriel
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas 13083970, Brazil
| |
Collapse
|
356
|
Hama T, Nagesh PK, Chowdhury P, Moore BM, Yallapu MM, Regner KR, Park F. DNA damage is overcome by TRIP13 overexpression during cisplatin nephrotoxicity. JCI Insight 2021; 6:139092. [PMID: 34806647 PMCID: PMC8663775 DOI: 10.1172/jci.insight.139092] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 10/13/2021] [Indexed: 11/17/2022] Open
Abstract
Cisplatin is a commonly used chemotherapeutic agent to treat a wide array of cancers that is frequently associated with toxic injury to the kidney due to oxidative DNA damage and perturbations in cell cycle progression leading to cell death. In this study, we investigated whether thyroid receptor interacting protein 13 (TRIP13) plays a central role in the protection of the tubular epithelia following cisplatin treatment by circumventing DNA damage. Following cisplatin treatment, double-stranded DNA repair pathways were inhibited using selective blockers to proteins involved in either homologous recombination or non-homologous end joining. This led to increased blood markers of acute kidney injury (AKI) (creatinine and neutrophil gelatinase–associated lipocalin), tubular damage, activation of DNA damage marker (γ-H2AX), elevated appearance of G2/M blockade (phosphorylated histone H3 Ser10 and cyclin B1), and apoptosis (cleaved caspase-3). Conditional proximal tubule–expressing Trip13 mice were observed to be virtually protected from the cisplatin nephrotoxicity by restoring most of the pathological phenotypes back toward normal conditions. Our findings suggest that TRIP13 could circumvent DNA damage in the proximal tubules during cisplatin injury and that TRIP13 may constitute a new therapeutic target in protecting the kidney from nephrotoxicants and reduce outcomes leading to AKI.
Collapse
Affiliation(s)
- Taketsugu Hama
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Prashanth Kb Nagesh
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
| | - Pallabita Chowdhury
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Bob M Moore
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Murali M Yallapu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA.,Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, Texas, USA
| | - Kevin R Regner
- Division of Nephrology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
357
|
Zazuli Z, de Jong C, Xu W, Vijverberg SJH, Masereeuw R, Patel D, Mirshams M, Khan K, Cheng D, Ordonez-Perez B, Huang S, Spreafico A, Hansen AR, Goldstein DP, de Almeida JR, Bratman SV, Hope A, Knox JJ, Wong RKS, Darling GE, Kitchlu A, van Haarlem SWA, van der Meer F, van Lindert ASR, ten Heuvel A, Brouwer J, Ross CJD, Carleton BC, Egberts TCG, Herder GJM, Deneer VHM, Maitland-van der Zee AH, Liu G. Association between Genetic Variants and Cisplatin-Induced Nephrotoxicity: A Genome-Wide Approach and Validation Study. J Pers Med 2021; 11:jpm11111233. [PMID: 34834585 PMCID: PMC8623115 DOI: 10.3390/jpm11111233] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/16/2022] Open
Abstract
This study aims to evaluate genetic risk factors for cisplatin-induced nephrotoxicity by investigating not previously studied genetic risk variants and further examining previously reported genetic associations. A genome-wide study (GWAS) was conducted in genetically estimated Europeans in a discovery cohort of cisplatin-treated adults from Toronto, Canada, followed by a candidate gene approach in a validation cohort from the Netherlands. In addition, previously reported genetic associations were further examined in both the discovery and validation cohorts. The outcome, nephrotoxicity, was assessed in two ways: (i) decreased estimated glomerular filtration rate (eGFR), calculated using the Chronic Kidney Disease Epidemiology Collaboration formula (CKD-EPI) and (ii) increased serum creatinine according to the Common Terminology Criteria for Adverse Events v4.03 for acute kidney injury (AKI-CTCAE). Four different Illumina arrays were used for genotyping. Standard quality control was applied for pre- and post-genotype imputation data. In the discovery cohort (n = 608), five single-nucleotide polymorphisms (SNPs) reached genome-wide significance. The A allele in rs4388268 (minor allele frequency = 0.23), an intronic variant of the BACH2 gene, was consistently associated with increased risk of cisplatin-induced nephrotoxicity in both definitions, meeting genome-wide significance (β = −8.4, 95% CI −11.4–−5.4, p = 3.9 × 10−8) for decreased eGFR and reaching suggestive association (OR = 3.9, 95% CI 2.3–6.7, p = 7.4 × 10−7) by AKI-CTCAE. In the validation cohort of 149 patients, this variant was identified with the same direction of effect (eGFR: β = −1.5, 95% CI −5.3–2.4, AKI-CTCAE: OR = 1.7, 95% CI 0.8–3.5). Findings of our previously published candidate gene study could not be confirmed after correction for multiple testing. Genetic predisposition of BACH2 (rs4388268) might be important in the development of cisplatin-induced nephrotoxicity, indicating opportunities for mechanistic understanding, tailored therapy and preventive strategies.
Collapse
Affiliation(s)
- Zulfan Zazuli
- Department of Respiratory Medicine, Academic Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands or (Z.Z.); (S.J.H.V.)
- Department of Pharmacology-Clinical Pharmacy, School of Pharmacy, Bandung Institute of Technology, Bandung 40132, Indonesia
| | - Corine de Jong
- Department of Clinical Pharmacy, St. Antonius Hospital, 3430 EM Nieuwegein, The Netherlands;
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (T.C.G.E.); (V.H.M.D.)
| | - Wei Xu
- Department of Biostatistics, Dalla Lana School of Public Health, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada;
| | - Susanne J. H. Vijverberg
- Department of Respiratory Medicine, Academic Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands or (Z.Z.); (S.J.H.V.)
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute of Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands;
| | - Devalben Patel
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (D.P.); (M.M.); (K.K.); (D.C.); (A.S.); (A.R.H.); (J.J.K.)
| | - Maryam Mirshams
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (D.P.); (M.M.); (K.K.); (D.C.); (A.S.); (A.R.H.); (J.J.K.)
| | - Khaleeq Khan
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (D.P.); (M.M.); (K.K.); (D.C.); (A.S.); (A.R.H.); (J.J.K.)
| | - Dangxiao Cheng
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (D.P.); (M.M.); (K.K.); (D.C.); (A.S.); (A.R.H.); (J.J.K.)
| | - Bayardo Ordonez-Perez
- Department of Laboratory Medicine and Pathology, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada;
| | - Shaohui Huang
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (S.H.); (S.V.B.); (A.H.); (R.K.S.W.)
- Department of Otolaryngology–Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (D.P.G.); (J.R.d.A.)
| | - Anna Spreafico
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (D.P.); (M.M.); (K.K.); (D.C.); (A.S.); (A.R.H.); (J.J.K.)
| | - Aaron R. Hansen
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (D.P.); (M.M.); (K.K.); (D.C.); (A.S.); (A.R.H.); (J.J.K.)
| | - David P. Goldstein
- Department of Otolaryngology–Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (D.P.G.); (J.R.d.A.)
| | - John R. de Almeida
- Department of Otolaryngology–Head and Neck Surgery, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (D.P.G.); (J.R.d.A.)
| | - Scott V. Bratman
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (S.H.); (S.V.B.); (A.H.); (R.K.S.W.)
| | - Andrew Hope
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (S.H.); (S.V.B.); (A.H.); (R.K.S.W.)
| | - Jennifer J. Knox
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (D.P.); (M.M.); (K.K.); (D.C.); (A.S.); (A.R.H.); (J.J.K.)
| | - Rebecca K. S. Wong
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (S.H.); (S.V.B.); (A.H.); (R.K.S.W.)
| | - Gail E. Darling
- Department of Thoracic Surgery, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada;
| | - Abhijat Kitchlu
- Department of Medicine, Nephrology, University Health Network, University of Toronto, Toronto, ON M5G 2M9, Canada;
| | | | - Femke van der Meer
- Department of Pulmonology, Diakonessenhuis, 3582 KE Utrecht, The Netherlands;
| | - Anne S. R. van Lindert
- Department of Pulmonology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands;
| | - Alexandra ten Heuvel
- Department of Pulmonology, Groene Hart Hospital, 2803 HH Gouda, The Netherlands;
| | - Jan Brouwer
- Department of Pulmonology, Rivierenland Hospital, 4002 WP Tiel, The Netherlands;
| | - Colin J. D. Ross
- British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; (C.J.D.R.); (B.C.C.)
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Bruce C. Carleton
- British Columbia Children’s Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; (C.J.D.R.); (B.C.C.)
- Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia, Vancouver, BC V1Y 1T3, Canada
- Pharmaceutical Outcomes Program, British Columbia Children’s Hospital, Vancouver, BC V5Z 4H4, Canada
| | - Toine C. G. Egberts
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (T.C.G.E.); (V.H.M.D.)
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Gerarda J. M. Herder
- Meander Medical Center, Department of Pulmonology, 3813 TZ Amersfoort, The Netherlands;
| | - Vera H. M. Deneer
- Department of Clinical Pharmacy, Division Laboratories, Pharmacy, and Biomedical Genetics, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands; (T.C.G.E.); (V.H.M.D.)
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Anke H. Maitland-van der Zee
- Department of Respiratory Medicine, Academic Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands or (Z.Z.); (S.J.H.V.)
- Correspondence: (A.H.M.-v.d.Z.); (G.L.); Tel.: +31-(0)20-566-8137 (A.H.M.-v.d.Z.); +416-946-4501 (ext. 3428) (G.L.)
| | - Geoffrey Liu
- Division of Medical Oncology and Hematology, Department of Medicine, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON M5G 2M9, Canada; (D.P.); (M.M.); (K.K.); (D.C.); (A.S.); (A.R.H.); (J.J.K.)
- Departments of Medical Biophysics, Pharmacology and Toxicology, and Epidemiology, Dalla Lana School of Public Health and University of Toronto, Toronto, ON M5T 3M7, Canada
- Correspondence: (A.H.M.-v.d.Z.); (G.L.); Tel.: +31-(0)20-566-8137 (A.H.M.-v.d.Z.); +416-946-4501 (ext. 3428) (G.L.)
| |
Collapse
|
358
|
Moreno-Gordaliza E, Marazuela MD, Pastor Ó, Lázaro A, Gómez-Gómez MM. Lipidomics Reveals Cisplatin-Induced Renal Lipid Alterations during Acute Kidney Injury and Their Attenuation by Cilastatin. Int J Mol Sci 2021; 22:ijms222212521. [PMID: 34830406 PMCID: PMC8622622 DOI: 10.3390/ijms222212521] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/07/2021] [Accepted: 11/17/2021] [Indexed: 12/02/2022] Open
Abstract
Nephrotoxicity is a major complication of cisplatin-based chemotherapy, leading to acute kidney injury in ca. 30% of patients, with no preventive intervention or treatment available for clinical use. Cilastatin has proved to exert a nephroprotective effect for cisplatin therapies in in vitro and in vivo models, having recently entered clinical trials. A deeper understanding at the molecular level of cisplatin-induced renal damage and the effect of potential protective agents could be key to develop successful nephroprotective therapies and to establish new biomarkers of renal damage and nephroprotection. A targeted lipidomics approach, using LC-MS/MS, was employed for the quantification of 108 lipid species (comprising phospholipids, sphingolipids, and free and esterified cholesterol) in kidney cortex and medulla extracts from rats treated with cisplatin and/or cilastatin. Up to 56 and 63 lipid species were found to be altered in the cortex and medulla, respectively, after cisplatin treatment. Co-treatment with cilastatin attenuated many of these lipid changes, either totally or partially with respect to control levels. Multivariate analysis revealed that lipid species can be used to discriminate renal damage and nephroprotection, with cholesterol esters being the most discriminating species, along with sulfatides and phospholipids. Potential diagnostic biomarkers of cisplatin-induced renal damage and cilastatin nephroprotection were also found.
Collapse
Affiliation(s)
- Estefanía Moreno-Gordaliza
- Department of Analytical Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.D.M.); (M.M.G.-G.)
- Correspondence:
| | - Maria Dolores Marazuela
- Department of Analytical Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.D.M.); (M.M.G.-G.)
| | - Óscar Pastor
- Servicio de Bioquímica Clínica, UCA-CCM, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain;
| | - Alberto Lázaro
- Renal Physiopathology Laboratory, Department of Nephrology, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain;
- Department of Physiology, School of Medicine, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - María Milagros Gómez-Gómez
- Department of Analytical Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, 28040 Madrid, Spain; (M.D.M.); (M.M.G.-G.)
| |
Collapse
|
359
|
Lee D, Lee SR, Park BJ, Song JH, Kim JK, Ko Y, Kang KS, Kim KH. Identification of Renoprotective Phytosterols from Mulberry ( Morus alba) Fruit against Cisplatin-Induced Cytotoxicity in LLC-PK1 Kidney Cells. PLANTS (BASEL, SWITZERLAND) 2021; 10:2481. [PMID: 34834844 PMCID: PMC8623081 DOI: 10.3390/plants10112481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/09/2021] [Accepted: 11/14/2021] [Indexed: 11/29/2022]
Abstract
The aim of this study was to explore the protective effects of bioactive compounds from the fruit of the mulberry tree (Morus alba L.) against cisplatin-induced apoptosis in LLC-PK1 pig kidney epithelial cells. Morus alba fruit is a well-known edible fruit commonly used in traditional folk medicine. Chemical investigation of M. alba fruit resulted in the isolation and identification of six phytosterols (1-6). Their structures were determined as 7-ketositosterol (1), stigmast-4-en-3β-ol-6-one (2), (3β,6α)-stigmast-4-ene-3,6-diol (3), stigmast-4-ene-3β,6β-diol (4), 7β-hydroxysitosterol 3-O-β-d-glucoside (5), and 7α-hydroxysitosterol 3-O-β-d-glucoside (6) by analyzing their physical and spectroscopic data as well as liquid chromatography/mass spectrometry data. All compounds displayed protective effects against cisplatin-induced LLC-PK1 cell damage, improving cisplatin-induced cytotoxicity to more than 80% of the control value. Compound 1 displayed the best effect at a relatively low concentration by inhibiting the percentage of apoptotic cells following cisplatin treatment. Its molecular mechanisms were identified using Western blot assays. Treatment of LLC-PK1 cells with compound 1 decreased the upregulated phosphorylation of p38 and c-Jun N-terminal kinase (JNK) following cisplatin treatment. In addition, compound 1 significantly suppressed cleaved caspase-3 in cisplatin-induced LLC-PK1 cells. Taken together, these findings indicated that cisplatin-induced apoptosis was significantly inhibited by compound 1 in LLC-PK1 cells, thereby supporting the potential of 7-ketositosterol (1) as an adjuvant candidate for treating cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Dahae Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea;
| | - Seoung Rak Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea;
| | - Bang Ju Park
- Department of Electronic Engineering, Gachon University, Seongnam 13120, Korea;
| | - Ji Hoon Song
- Jeju Institute of Korean Medicine, Jeju 63309, Korea;
| | - Jung Kyu Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Korea;
| | - Yuri Ko
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA;
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea;
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Korea;
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA;
| |
Collapse
|
360
|
Abouzed TK, Sherif EAE, Barakat MES, Sadek KM, Aldhahrani A, Nasr NE, Eldomany E, Khailo K, Dorghamm DA. Assessment of gentamicin and cisplatin-induced kidney damage mediated via necrotic and apoptosis genes in albino rats. BMC Vet Res 2021; 17:350. [PMID: 34784920 PMCID: PMC8594120 DOI: 10.1186/s12917-021-03023-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 09/15/2021] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Gentamicin (GM) is a low-cost, low-resistance antibiotic commonly used to treat gram-negative bacterial diseases. Cisplatin (Csp) is a platinum-derived anti-neoplastic agent. This experiment aimed to identify the early signs of gentamicin and cisplatin-induced nephrotoxicity in rats. Thirty Wistar rats were divided into three groups of 10: a control group, which received no treatment; a gentamicin group administered by a dose of (100 mg/kg, IP) for 7 consecutive days, and a cisplatin group was administered intraperitoneal in a dose of (1.5 mg/kg body weight) repeated twice a week for 3 weeks. RESULTS Both experimental groups exhibited increased levels of creatinine, urea, and uric acid, with the cisplatin-treated group showing higher levels than the gentamicin group. Experimental groups also exhibited significantly increased Malondialdehyde (MDA), reduced glutathione (GSH), and glutathione peroxidase (GSH-Px) with more pronounced effects in the cisplatin-treated group. Further, both experimental groups exhibited significant up-regulation of Tumor Necrosis Factor α (TNF-α), caspase-3, and Bax and down regulation of Bcl-2. CONCLUSION These findings confirm the use of necrotic, apoptotic genes as early biomarkers in the detection of tubular kidney damage. Further, cisplatin was shown to have a greater nephrotoxic effect than gentamicin; therefore, its use should be constrained accordingly when co-administered with gentamicin.
Collapse
Affiliation(s)
- Tarek Kamal Abouzed
- Biochemistry Department, Faculty of Veterinary Medicine Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Eman Abd Elrahman Sherif
- Biochemistry Department, Faculty of Veterinary Medicine Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Mohamed El Sayed Barakat
- Biochemistry Unit, Animal Health Research Institute, Kafrelsheikh branch. Agricultural Research Center (ARC), Kafrelsheikh, Egypt.
| | - Kadry Mohamed Sadek
- Biochemistry Department, Faculty of Veterinary Medicine Damanhour University, Damanhour, Egypt
| | - Adil Aldhahrani
- Clinical laboratory science Department, Turabah University College, Taif University, Taif, Saudi Arabia
| | - Nasr Elsayed Nasr
- Biochemistry Department, Faculty of Veterinary Medicine Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Ehab Eldomany
- Department of Biotechnology and Life science, Faculty of Postgraduate Studies for Advanced Science Beni-suef University, Beni-suef, Egypt
| | - Khaled Khailo
- Biochemistry Department, Faculty of Veterinary Medicine Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Doaa Abdallha Dorghamm
- Biochemistry Department, Faculty of Veterinary Medicine Kafrelsheikh University, Kafrelsheikh, Egypt
| |
Collapse
|
361
|
Naushad M, Urooj M, Ahmad T, Husain GM, Kazmi MH, Zakir M. Nephroprotective effect of Apium graveolens L. against Cisplatin-induced nephrotoxicity. J Ayurveda Integr Med 2021; 12:607-615. [PMID: 34774409 PMCID: PMC8642661 DOI: 10.1016/j.jaim.2021.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 06/06/2021] [Accepted: 06/08/2021] [Indexed: 11/23/2022] Open
Abstract
Background Cisplatin is extensively used in treating cancers, and its primary side-effect is nephrotoxicity. It accumulates in proximal convoluted tubules where it promotes cellular damage by oxidative stress, apoptosis, and inflammation, etc. In Unani medicine, Tukhm-e-Karafs(Apium graveolens L.) (TK) is mentioned in the literature to manage various kidney ailments due to its diuretic and deobstruent activities. Objective To investigate the nephroprotective effects of powder of TK in Cisplatin-induced nephrotoxicity in an animal model and to validate the Unani claim of its nephroprotective action. Material and methods In curative protocol, cisplatin (5 mg/kg body weight i.p) was administered on day one and powder of TK (500 and 1000 mg/kg p.o.) from the sixth day onwards for ten days. TK (500 and 1000 mg/kg p.o.) was given for ten days and Cisplatin (5 mg/kg body weight i.p) on day 11 in the protective model. At the end of the study, all the animals were sacrificed, and renal biochemical parameters were determined. KIM-1 level was also investigated in the kidney homogenate in conjunction with histopathological inspection of kidney tissues. Results Significant increase in serum creatinine and BUN, presence of mononuclear cell infiltration, tubular dilation and vacuolation in renal histopathology, and increased KIM-1 level confirmed the nephrotoxicity due to Cisplatin. TK's administration protects the kidney as suggested by the changes in biochemical renal function, decreased level of KIM-1, and improvement in histopathological changes. Conclusion The result advocated that TK prevented renal injury and maintained normal renal function in both models. It may be due to improved clearance of Cisplatin from kidney tubules and reduction in reactive oxygen species (ROS) produced by the inflammatory response.
Collapse
Affiliation(s)
- Mohd Naushad
- Department of Ilmul Advia, National Research Institute of Unani Medicine for Skin Disorders (NRIUMSD), Hyderabad, India
| | - Mohd Urooj
- National Research Institute of Unani Medicine for Skin Disorders (NRIUMSD), Hyderabad, India
| | - Tasleem Ahmad
- National Research Institute of Unani Medicine for Skin Disorders (NRIUMSD), Hyderabad, India
| | - Gulam Mohammed Husain
- National Research Institute of Unani Medicine for Skin Disorders (NRIUMSD), Hyderabad, India
| | - Munawwar Husain Kazmi
- Department of Ilmul Advia, National Research Institute of Unani Medicine for Skin Disorders (NRIUMSD), Hyderabad, India
| | - Mohammad Zakir
- Department of Ilmul Advia, National Research Institute of Unani Medicine for Skin Disorders (NRIUMSD), Hyderabad, India.
| |
Collapse
|
362
|
Tian X, Yang F, Li F, Ran L, Chang J, Li J, Hong W, Shan L, Du Y, Hu L, Mei F, He M, Li Y, Wang H, Zuo K, Zhou B, Chen S, Mao W. A Comparison of Different Schemes of Neoadjuvant Chemotherapy Followed by Concurrent Chemotherapy and Radiotherapy for Locally Advanced Cervical Cancer: A Retrospective Study. Cancer Manag Res 2021; 13:8307-8316. [PMID: 34764693 PMCID: PMC8572729 DOI: 10.2147/cmar.s328309] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/21/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose To examine the clinical significance of unoperated cervical cancer patients treated with different neoadjuvant chemotherapy (NACT) schemes followed by concurrent chemotherapy and radiotherapy (CCRT). Methods This retrospective analysis included women with locally advanced cervical cancer treated with NACT-CCRT between September 2011 and September 2014. Neoadjuvant chemotherapy included paclitaxel plus cisplatin (TP group; 62 patients) or paclitaxel plus loplatin (TL group; 58 patients), which were administered three weekly, and cisplatin or loplatin, which were administered weekly for synchronous chemotherapy. External beam radiation therapy (50.4–56.35 Gy/28 f, 180–215 cGy/f, 5 f/w) was followed by intracavitary brachytherapy (5 Gy per fraction, mostly 5 fractions, Ir192 based). Results One hundred twenty women were included in the analysis. The complete/partial response rate was 99.2% after treatment. The one-year, three-year, and five-year survival rates were 99.2%, 82.5%, and 70.8%, respectively. In the TP and TL groups, the three-year and five-year survival rates were 85.5% vs 77.6% and 75.8% vs 65.5%, respectively, with no significant difference. The 5-year overall survival (OS) rates between patients with stage IIB and stage IIIB disease were not significantly different (69.2% vs 64.7%). In the TP group, grade 3 or 4 digestive reactions were more frequent than those in the TL group. Leukopenia, neutropenia, and thrombocytopenia were more common in the TL group. No significant difference was found in anemia, radiation enteritis, radiation proctitis, or radiation cystitis between the groups. Conclusion Lobaplatin may be used as an alternative drug for patients with severe digestive system reactions or contraindications to cisplatin, but hematological toxicity must be considered, particularly in dose-intensive schemes. Neoadjuvant chemotherapy followed by concurrent chemotherapy and radiotherapy (NACT-CCRT) warrants further prospective study in cervical cancer patients with a wide range of tumor invasion (eg, mass size ≥5 cm or stage IIIB).
Collapse
Affiliation(s)
- Xue Tian
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Feiyue Yang
- Department of Gynecologic Oncology, Guizhou Provincial People's Hospital, Guiyang, 550004, People's Republic of China
| | - Fenghu Li
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Li Ran
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Jianying Chang
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Jiehui Li
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Wei Hong
- Department of Radiotherapy, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Lang Shan
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Yanjun Du
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Lili Hu
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Fan Mei
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Mingyuan He
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Yongxia Li
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Heran Wang
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Kai Zuo
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Bo Zhou
- Surgical Department of Gynecological Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Shuying Chen
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Wanli Mao
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| |
Collapse
|
363
|
Hu X, Ma Z, Wen L, Li S, Dong Z. Autophagy in Cisplatin Nephrotoxicity during Cancer Therapy. Cancers (Basel) 2021; 13:5618. [PMID: 34830772 PMCID: PMC8616020 DOI: 10.3390/cancers13225618] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/23/2021] [Accepted: 11/04/2021] [Indexed: 12/12/2022] Open
Abstract
Cisplatin is a widely used chemotherapeutic agent but its clinical use is often limited by nephrotoxicity. Autophagy is a lysosomal degradation pathway that removes protein aggregates and damaged or dysfunctional cellular organelles for maintaining cell homeostasis. Upon cisplatin exposure, autophagy is rapidly activated in renal tubule cells to protect against acute cisplatin nephrotoxicity. Mechanistically, the protective effect is mainly related to the clearance of damaged mitochondria via mitophagy. The role and regulation of autophagy in chronic kidney problems after cisplatin treatment are currently unclear, despite the significance of research in this area. In cancers, autophagy may prevent tumorigenesis, but autophagy may reduce the efficacy of chemotherapy by protecting cancer cells. Future research should focus on developing drugs that enhance the anti-tumor effects of cisplatin while protecting kidneys during cisplatin chemotherapy.
Collapse
Affiliation(s)
- Xiaoru Hu
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (X.H.); (L.W.); (S.L.)
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Zhengwei Ma
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Lu Wen
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (X.H.); (L.W.); (S.L.)
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Siyao Li
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (X.H.); (L.W.); (S.L.)
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Zheng Dong
- Hunan Key Laboratory of Kidney Disease and Blood Purification, Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (X.H.); (L.W.); (S.L.)
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA;
- Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| |
Collapse
|
364
|
McMahon KR, Chui H, Rassekh SR, Schultz KR, Blydt-Hansen TD, Mammen C, Pinsk M, Cuvelier GDE, Carleton BC, Tsuyuki RT, Ross CJ, Devarajan P, Huynh L, Yordanova M, Crépeau-Hubert F, Wang S, Cockovski V, Palijan A, Zappitelli M. Urine Neutrophil Gelatinase-Associated Lipocalin and Kidney Injury Molecule-1 to Detect Pediatric Cisplatin-Associated Acute Kidney Injury. KIDNEY360 2021; 3:37-50. [PMID: 35368557 PMCID: PMC8967607 DOI: 10.34067/kid.0004802021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/29/2021] [Indexed: 01/12/2023]
Abstract
Background Few studies have described associations between the AKI biomarkers urinary neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1) with AKI in cisplatin-treated children. We aimed to describe excretion patterns of urine NGAL and KIM-1 and associations with AKI in children receiving cisplatin. Methods Participants (n=159) were enrolled between 2013 and 2017 in a prospective cohort study conducted in 12 Canadian pediatric hospitals. Participants were evaluated at early cisplatin infusions (at first or second cisplatin cycle) and late cisplatin infusions (last or second-to-last cycle). Urine NGAL and KIM-1 were measured (1) pre-cisplatin infusion, (2) post-infusion (morning after), and (3) at hospital discharge at early and late cisplatin infusions. Primary outcome: AKI defined by serum creatinine rise within 10 days post-cisplatin, on the basis of Kidney Disease Improving Global Outcomes guidelines criteria (stage 1 or higher). Results Of 159 children, 156 (median [interquartile range (IQR)] age: 5.8 [2.4-12.0] years; 78 [50%] female) had biomarker data available at early cisplatin infusions and 127 had data at late infusions. Forty six of the 156 (29%) and 22 of the 127 (17%) children developed AKI within 10 days of cisplatin administration after early and late infusions, respectively. Urine NGAL and KIM-1 concentrations were significantly higher in patients with versus without AKI (near hospital discharge of late cisplatin infusion, median [IQR] NGAL levels were 76.1 [10.0-232.7] versus 14.9 [5.4-29.7] ng/mg creatinine; KIM-1 levels were 4415 [2083-9077] versus 1049 [358-3326] pg/mg creatinine; P<0.01). These markers modestly discriminated for AKI (area under receiver operating characteristic curve [AUC-ROC] range: NGAL, 0.56-0.72; KIM-1, 0.48-0.75). Biomarker concentrations were higher and better discriminated for AKI at late cisplatin infusions (AUC-ROC range, 0.54-0.75) versus early infusions (AUC-ROC range, 0.48-0.65). Conclusions Urine NGAL and KIM-1 were modest at discriminating for cisplatin-associated AKI. Further research is needed to determine clinical utility and applicability of these markers and associations with late kidney outcomes.
Collapse
Affiliation(s)
- Kelly R. McMahon
- Division of Nephrology, Department of Pediatrics, Research Institute of the McGill University Health Centre, McGill University Health Centre, Montreal Children’s Hospital, Montreal, Quebec, Canada,Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | - Hayton Chui
- Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada,Faculty of Health Sciences, McMaster Health Sciences Centre, McMaster University, Hamilton, Ontario, Canada
| | - Shahrad Rod Rassekh
- Division of Hematology/Oncology/Bone Marrow Transplantation, Department of Pediatrics, University of British Columbia, British Columbia Children’s Hospital, Vancouver, British Columbia, Canada
| | - Kirk R. Schultz
- Division of Hematology/Oncology/Bone Marrow Transplantation, Department of Pediatrics, University of British Columbia, British Columbia Children’s Hospital, Vancouver, British Columbia, Canada
| | - Tom D. Blydt-Hansen
- Division of Pediatric Nephrology, Department of Pediatrics, University of British Columbia, British Columbia Children’s Hospital, Vancouver, British Columbia, Canada
| | - Cherry Mammen
- Division of Pediatric Nephrology, Department of Pediatrics, University of British Columbia, British Columbia Children’s Hospital, Vancouver, British Columbia, Canada
| | - Maury Pinsk
- Department of Pediatrics and Child Health, Section of Pediatric Nephrology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Geoffrey D. E. Cuvelier
- Division of Pediatric Oncology-Hematology-BMT, Department of Pediatrics and Child Health, University of Manitoba, CancerCare Manitoba, Winnipeg, Manitoba, Canada
| | - Bruce C. Carleton
- Division of Translational Therapeutics, Department of Pediatrics, University of British Columbia and BC Children’s Hospital and Research Institute, Vancouver, British Columbia, Canada
| | - Ross T. Tsuyuki
- EPICORE Centre, Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Colin J.D. Ross
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Prasad Devarajan
- Division of Nephrology and Hypertension, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Louis Huynh
- Faculty of Health Sciences, Queen’s University, Kingston, Ontario, Canada
| | - Mariya Yordanova
- Faculty of Medicine and Health Sciences, Sherbrooke University, Sherbrooke, Quebec, Canada
| | - Frédérik Crépeau-Hubert
- Division of Nephrology, Department of Pediatrics, Research Institute of the McGill University Health Centre, McGill University Health Centre, Montreal Children’s Hospital, Montreal, Quebec, Canada
| | - Stella Wang
- Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada
| | - Vedran Cockovski
- Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada
| | - Ana Palijan
- Division of Nephrology, Department of Pediatrics, Research Institute of the McGill University Health Centre, McGill University Health Centre, Montreal Children’s Hospital, Montreal, Quebec, Canada
| | - Michael Zappitelli
- Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada,Department of Pediatrics, Division of Nephrology, Toronto Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
365
|
Thongnuanjan P, Soodvilai S, Fongsupa S, Thipboonchoo N, Chabang N, Munyoo B, Tuchinda P, Soodvilai S. Panduratin A Derivative Protects against Cisplatin-Induced Apoptosis of Renal Proximal Tubular Cells and Kidney Injury in Mice. Molecules 2021; 26:6642. [PMID: 34771049 PMCID: PMC8588142 DOI: 10.3390/molecules26216642] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/26/2021] [Accepted: 10/28/2021] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Panduratin A is a bioactive cyclohexanyl chalcone exhibiting several pharmacological activities, such as anti-inflammatory, anti-oxidative, and anti-cancer activities. Recently, the nephroprotective effect of panduratin A in cisplatin (CDDP) treatment was revealed. The present study examined the potential of certain compounds derived from panduratin A to protect against CDDP-induced nephrotoxicity. METHODS Three derivatives of panduratin A (DD-217, DD-218, and DD-219) were semi-synthesized from panduratin A. We investigated the effects and corresponding mechanisms of the derivatives of panduratin A for preventing nephrotoxicity of CDDP in both immortalized human renal proximal tubular cells (RPTEC/TERT1 cells) and mice. RESULTS Treating the cell with 10 µM panduratin A significantly reduced the viability of RPTEC/TERT1 cells compared to control (panduratin A: 72% ± 4.85%). Interestingly, DD-217, DD-218, and DD-219 at the same concentration did not significantly affect cell viability (92% ± 8.44%, 90% ± 7.50%, and 87 ± 5.2%, respectively). Among those derivatives, DD-218 exhibited the most protective effect against CDDP-induced renal proximal tubular cell apoptosis (control: 57% ± 1.23%; DD-218: 19% ± 10.14%; DD-219: 33% ± 14.06%). The cytoprotective effect of DD-218 was mediated via decreases in CDDP-induced mitochondria dysfunction, intracellular reactive oxygen species (ROS) generation, activation of ERK1/2, and cleaved-caspase 3 and 7. In addition, DD-218 attenuated CDDP-induced nephrotoxicity by a decrease in renal injury and improved in renal dysfunction in C57BL/6 mice. Importantly, DD-218 did not attenuate the anti-cancer efficacy of CDDP in non-small-cell lung cancer cells or colon cancer cells. CONCLUSIONS This finding suggests that DD-218, a derivative of panduratin A, holds promise as an adjuvant therapy in patients receiving CDDP.
Collapse
Affiliation(s)
- Penjai Thongnuanjan
- Toxicology Graduate Program, Multidisciplinary Unit, Faculty of Science, Mahidol University, Rama VI Road, Ratchathewi, Bangkok 10400, Thailand;
- Research Center of Transport Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Ratchathewi, Bangkok 10400, Thailand;
| | - Sirima Soodvilai
- Department of Pharmaceutical Technology, College of Pharmacy, Rangsit University, Pathumthani 12000, Thailand;
| | - Somsak Fongsupa
- Department of Medical Technology, Faculty of Allied Health Science, Thammasat University, Pathumthani 12121, Thailand;
| | - Natechanok Thipboonchoo
- Research Center of Transport Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Ratchathewi, Bangkok 10400, Thailand;
| | - Napason Chabang
- School of Bioinnovation and Bio-Based Product Intelligence, Faculty of Science, Mahidol University, Rama VI Road, Ratchathewi, Bangkok 10400, Thailand;
| | - Bamroong Munyoo
- Excellent Center for Drug Discovery, Mahidol University, Rama VI Road, Ratchathewi, Bangkok 10400, Thailand; (B.M.); (P.T.)
| | - Patoomratana Tuchinda
- Excellent Center for Drug Discovery, Mahidol University, Rama VI Road, Ratchathewi, Bangkok 10400, Thailand; (B.M.); (P.T.)
| | - Sunhapas Soodvilai
- Toxicology Graduate Program, Multidisciplinary Unit, Faculty of Science, Mahidol University, Rama VI Road, Ratchathewi, Bangkok 10400, Thailand;
- Research Center of Transport Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Rama VI Road, Ratchathewi, Bangkok 10400, Thailand;
- Excellent Center for Drug Discovery, Mahidol University, Rama VI Road, Ratchathewi, Bangkok 10400, Thailand; (B.M.); (P.T.)
| |
Collapse
|
366
|
Lee D, Kang KB, Hwang GS, Choi YK, Kim TK, Kang KS. Antioxidant and Anti-Inflammatory Effects of 3-Dehydroxyceanothetric Acid 2-Methyl Ester Isolated from Ziziphus jujuba Mill. against Cisplatin-Induced Kidney Epithelial Cell Death. Biomolecules 2021; 11:1614. [PMID: 34827612 PMCID: PMC8615384 DOI: 10.3390/biom11111614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 11/17/2022] Open
Abstract
Cisplatin is a platinum-based chemotherapeutic agent for treating solid tumors; however, it presents a risk factor for nephropathy. In the present study, we investigated the antioxidant and anti-inflammatory effects of 3-dehydroxyceanothetric acid 2-methyl ester (3DC2ME) isolated from Ziziphus jujuba Mill. in LLC-PK1 cells following cisplatin-induced cytotoxicity. These cells were exposed to 3DC2ME for 2 h, followed by treatment with cisplatin for 24 h. The treated cells were subjected to cell viability analysis using the Ez-Cytox assay. Reactive oxygen species (ROS) were detected via 2', 7'- dichlorodihydrofluorescein diacetate (DCFH-DA) staining. In addition, western blotting and fluorescent immunostaining were performed to evaluate protein expressions related to oxidative stress and inflammation pathways. Pretreatment with 3DC2ME protected LLC-PK1 cells from cisplatin-induced cytotoxicity and oxidative stress. In addition, pretreatment with 3DC2ME upregulated heme oxygenase 1 (HO-1) via the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in the cisplatin-treated LLC-PK1 cells. Furthermore, the increase in the expressions of IκB kinase α/β (IKKα/β), inhibitor of kappa B alpha (IκBα), nuclear factor kappa B (NF-κB), inducible nitric oxide synthase (iNOS), and cyclooxygenase-2 (COX-2) in these cells was inhibited. These results provide basic scientific evidence for understanding the antioxidant and anti-inflammatory effects of 3DC2ME isolated from Z. jujuba against cisplatin-induced kidney epithelial cell death.
Collapse
Affiliation(s)
- Dahae Lee
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea; (D.L.); (G.S.H.); (Y.-K.C.)
| | - Kyo Bin Kang
- College of Pharmacy, Sookmyung Women’s University, Seoul 04310, Korea;
| | - Gwi Seo Hwang
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea; (D.L.); (G.S.H.); (Y.-K.C.)
| | - You-Kyoung Choi
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea; (D.L.); (G.S.H.); (Y.-K.C.)
| | - Tae Kon Kim
- College of Science & Engineering, Jungwon University, Chungbuk 28024, Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Korea; (D.L.); (G.S.H.); (Y.-K.C.)
| |
Collapse
|
367
|
Nozhat Z, Heydarzadeh S, Memariani Z, Ahmadi A. Chemoprotective and chemosensitizing effects of apigenin on cancer therapy. Cancer Cell Int 2021; 21:574. [PMID: 34715860 PMCID: PMC8555304 DOI: 10.1186/s12935-021-02282-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 10/20/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Therapeutic resistance to radiation and chemotherapy is one of the major obstacles in cancer treatment. Although synthetic radiosensitizers are pragmatic solution to enhance tumor sensitivity, they pose concerns of toxicity and non-specificity. In the last decades, scientists scrutinized novel plant-derived radiosensitizers and chemosensitizers, such as flavones, owing to their substantial physiological effects like low toxicity and non-mutagenic properties on the human cells. The combination therapy with apigenin is potential candidate in cancer therapeutics. This review explicates the combinatorial strategies involving apigenin to overcome drug resistance and boost the anti-cancer properties. METHODS We selected full-text English papers on international databases like PubMed, Web of Science, Google Scholar, Scopus, and ScienceDirect from 1972 up to 2020. The keywords included in the search were: Apigenin, Chemoprotective, Chemosensitizing, Side Effects, and Molecular Mechanisms. RESULTS In this review, we focused on combination therapy, particularly with apigenin augmenting the anti-cancer effects of chemo drugs on tumor cells, reduce their side effects, subdue drug resistance, and protect healthy cells. The reviewed research data implies that these co-therapies exhibited a synergistic effect on various cancer cells, where apigenin sensitized the chemo drug through different pathways including a significant reduction in overexpressed genes, AKT phosphorylation, NFκB, inhibition of Nrf2, overexpression of caspases, up-regulation of p53 and MAPK, compared to the monotherapies. Meanwhile, contrary to the chemo drugs alone, combined treatments significantly induced apoptosis in the treated cells. CONCLUSION Briefly, our analysis proposed that the combination therapies with apigenin could suppress the unwanted toxicity of chemotherapeutic agents. It is believed that these expedient results may pave the path for the development of drugs with a high therapeutic index. Nevertheless, human clinical trials are a prerequisite to consider the potential use of apigenin in the prevention and treatment of various cancers. Conclusively, the clinical trials to comprehend the role of apigenin as a chemoprotective agent are still in infancy.
Collapse
Affiliation(s)
- Zahra Nozhat
- Institute of Smart Biomedical Materials, School of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018 China
- Cellular and Molecular Endocrine Research Center, Research Institute of Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shabnam Heydarzadeh
- Cellular and Molecular Endocrine Research Center, Research Institute of Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biochemistry, School of Biological Sciences, Falavarjan Branch Islamic Azad University, Isfahan, Iran
| | - Zahra Memariani
- Traditional Medicine and History of Medical Sciences Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Amirhossein Ahmadi
- Pharmaceutical Sciences Research Center, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
368
|
Abadi AJ, Mirzaei S, Mahabady MK, Hashemi F, Zabolian A, Hashemi F, Raee P, Aghamiri S, Ashrafizadeh M, Aref AR, Hamblin MR, Hushmandi K, Zarrabi A, Sethi G. Curcumin and its derivatives in cancer therapy: Potentiating antitumor activity of cisplatin and reducing side effects. Phytother Res 2021; 36:189-213. [PMID: 34697839 DOI: 10.1002/ptr.7305] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/03/2021] [Accepted: 09/25/2021] [Indexed: 12/12/2022]
Abstract
Curcumin is a phytochemical isolated from Curcuma longa with potent tumor-suppressor activity, which has shown significant efficacy in pre-clinical and clinical studies. Curcumin stimulates cell death, triggers cycle arrest, and suppresses oncogenic pathways, thereby suppressing cancer progression. Cisplatin (CP) stimulates DNA damage and apoptosis in cancer chemotherapy. However, CP has adverse effects on several organs of the body, and drug resistance is frequently observed. The purpose of the present review is to show the function of curcumin in decreasing CP's adverse impacts and improving its antitumor activity. Curcumin administration reduces ROS levels to prevent apoptosis in normal cells. Furthermore, curcumin can inhibit inflammation via down-regulation of NF-κB to maintain the normal function of organs. Curcumin and its nanoformulations can reduce the hepatoxicity, neurotoxicity, renal toxicity, ototoxicity, and cardiotoxicity caused by CP. Notably, curcumin potentiates CP cytotoxicity via mediating cell death and cycle arrest. Besides, curcumin suppresses the STAT3 and NF-ĸB as tumor-promoting pathways, to enhance CP sensitivity and prevent drug resistance. The targeted delivery of curcumin and CP to tumor cells can be mediated nanostructures. In addition, curcumin derivatives are also able to reduce CP-mediated side effects, and increase CP cytotoxicity against various cancer types.
Collapse
Affiliation(s)
- Asal Jalal Abadi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Farid Hashemi
- Department of Comparative Biosciences, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fardin Hashemi
- School of Rehabilitation, Department of Physical Therapy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Pourya Raee
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahin Aghamiri
- Student Research Committee, Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Tuzla, Turkey.,Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA.,Vice President at Translational Sciences, Xsphera Biosciences Inc, Boston, Massachusetts, USA
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa.,Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Istanbul, Turkey.,Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer, Istanbul, Turkey
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
369
|
Yamashita N, Nakai K, Nakata T, Nakamura I, Kirita Y, Matoba S, Humphreys BD, Tamagaki K, Kusaba T. Cumulative DNA damage by repeated low-dose cisplatin injection promotes the transition of acute to chronic kidney injury in mice. Sci Rep 2021; 11:20920. [PMID: 34686727 PMCID: PMC8536734 DOI: 10.1038/s41598-021-00392-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/12/2021] [Indexed: 12/14/2022] Open
Abstract
Cisplatin is a commonly used anticancer drug, but nephrotoxicity is a dose-limiting adverse effect. Recent experimental and clinical observations have demonstrated that multiple injections of cisplatin induce the transition to chronic kidney disease; however, the underlying mechanisms remain unclear. We found that multiple injections of higher doses of cisplatin in a shorter interval affected the severity of kidney injury, causing kidney fibrosis to develop at a later time point. An additional injection of cisplatin during the recovery period after a prior injury, when proximal tubule epithelia are actively proliferating, induced substantial tubular injury by inducing more severe DNA damage than that induced by a single injection. Lineage tracing analysis of proximal tubular epithelia demonstrated that the tubular epithelia that underwent multiple rounds of cell division after multiple injections of cisplatin existed at the chronic phase, and these populations often expressed vcam1 + , suggesting the induction of proinflammatory failed-repair tubular epithelia. Our study revealed that as cisplatin exerts cytotoxic effects on actively proliferating cells, additional cisplatin injections before the completion of tubular repair exacerbates kidney injury through cumulative DNA damage. Appropriate both the setting of dosage and dosing intervals, with careful monitoring, are essential to prevent nephrotoxicity of repeated cisplatin treatment in cancer patients.
Collapse
Affiliation(s)
- Noriyuki Yamashita
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan.,Institute of Experimental Medicine and Systems Biology, RWTH Aachen University, Aachen, Germany
| | - Kunihiro Nakai
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Tomohiro Nakata
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Itaru Nakamura
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yuhei Kirita
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Satoaki Matoba
- Department of Cardiovascular Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Benjamin D Humphreys
- Division of Nephrology, Washington University School of Medicine in St. Louis, St. Louis, USA
| | - Keiichi Tamagaki
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Tetsuro Kusaba
- Department of Nephrology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto, 602-8566, Japan.
| |
Collapse
|
370
|
Budu A, Freitas-Lima LC, Arruda ACD, Perilhão MS, Barrera-Chimal J, Araújo RC, Estrela GR. Renal fibrosis due to multiple cisplatin treatment is exacerbated by kinin B1 receptor antagonism. Braz J Med Biol Res 2021; 54:e11353. [PMID: 34669782 PMCID: PMC8521536 DOI: 10.1590/1414-431x2021e11353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/13/2021] [Indexed: 11/22/2022] Open
Abstract
Cisplatin is a widely used chemotherapeutic drug, but its side effects are a major limiting factor. Nephrotoxicity occurs in one third of patients undergoing cisplatin treatment. The acute tubular injury caused by cisplatin often leads to a defective repair process, which translates into chronic renal disorders. In this way, cisplatin affects tubular cells, and maladaptive tubules regeneration will ultimately result in tubulointerstitial fibrosis. Kinins are well known for being important peptides in the regulation of inflammatory stimuli, and kinin B1 receptor deficiency and antagonism have been shown to be beneficial against acute cisplatin nephrotoxicity. This study aimed to analyze the effects of kinin B1 receptor deletion and antagonism against repeated cisplatin-induced chronic renal dysfunction and fibrosis. Both the deletion and the antagonism of B1 receptor exacerbated cisplatin-induced chronic renal dysfunction. Moreover, the inhibition of B1 receptor increased tubular injury and tubulointerstitial fibrosis after repeated treatment with cisplatin. The balance between M1/M2 macrophage polarization plays an important role in renal fibrosis. Kinin B1 receptor antagonism had no impact on M1 markers when compared to cisplatin. However, YM1, an M2 marker and an important molecule for the wound healing process, was decreased in mice treated with kinin B1 receptor antagonist, compared to cisplatin alone. Endothelin-1 levels were also increased in mice with B1 receptor inhibition. This study showed that kinin B1 receptor inhibition exacerbated cisplatin-induced chronic renal dysfunction and fibrosis, associated with reduced YM1 M2 marker expression, thus possibly affecting the wound healing process.
Collapse
Affiliation(s)
- A Budu
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - L C Freitas-Lima
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - A C de Arruda
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, SP, Brasil.,Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - M S Perilhão
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, SP, Brasil.,Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil.,Faculdade de Educação Física, Universidade Santo Amaro, São Paulo, SP, Brasil
| | - J Barrera-Chimal
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Unidad de Investigación UNAM-INC, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City, Mexico
| | - R C Araújo
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, SP, Brasil.,Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| | - G R Estrela
- Disciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brasil.,Disciplina de Hematologia e Hematoterapia, Departamento de Oncologia Clínica e Experimental, Universidade Federal de São Paulo, São Paulo, SP, Brasil
| |
Collapse
|
371
|
Alam W, Rocca C, Khan H, Hussain Y, Aschner M, De Bartolo A, Amodio N, Angelone T, Cheang WS. Current Status and Future Perspectives on Therapeutic Potential of Apigenin: Focus on Metabolic-Syndrome-Dependent Organ Dysfunction. Antioxidants (Basel) 2021; 10:antiox10101643. [PMID: 34679777 PMCID: PMC8533599 DOI: 10.3390/antiox10101643] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Metabolic syndrome and its associated disorders such as obesity, insulin resistance, atherosclerosis and type 2 diabetes mellitus are globally prevalent. Different molecules showing therapeutic potential are currently available for the management of metabolic syndrome, although their efficacy has often been compromised by their poor bioavailability and side effects. Studies have been carried out on medicinal plant extracts for the treatment and prevention of metabolic syndrome. In this regard, isolated pure compounds have shown promising efficacy for the management of metabolic syndrome, both in preclinical and clinical settings. Apigenin, a natural bioactive flavonoid widely present in medicinal plants, functional foods, vegetables and fruits, exerts protective effects in models of neurological disorders and cardiovascular diseases and most of these effects are attributed to its antioxidant action. Various preclinical and clinical studies carried out so far show a protective effect of apigenin against metabolic syndrome. Herein, we provide a comprehensive review on both in vitro and in vivo evidence related to the promising antioxidant role of apigenin in cardioprotection, neuroprotection and renoprotection, and to its beneficial action in metabolic-syndrome-dependent organ dysfunction. We also provide evidence on the potential of apigenin in the prevention and/or treatment of metabolic syndrome, analysing the potential and limitation of its therapeutic use.
Collapse
Affiliation(s)
- Waqas Alam
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan;
| | - Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences (Di.B.E.S.T.), University of Calabria, 87036 Rende, Italy; (C.R.); (A.D.B.)
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan;
- Correspondence: or (H.K.); (N.A.); (T.A.)
| | - Yaseen Hussain
- College of Pharmaceutical Sciences, Soochow University, Suzhou 221400, China;
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY 10461, USA;
| | - Anna De Bartolo
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences (Di.B.E.S.T.), University of Calabria, 87036 Rende, Italy; (C.R.); (A.D.B.)
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
- Correspondence: or (H.K.); (N.A.); (T.A.)
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences (Di.B.E.S.T.), University of Calabria, 87036 Rende, Italy; (C.R.); (A.D.B.)
- National Institute of Cardiovascular Research I.N.R.C., 40126 Bologna, Italy
- Correspondence: or (H.K.); (N.A.); (T.A.)
| | - Wai San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China;
| |
Collapse
|
372
|
Hydration with Mannitol and Dextrose May Promote Cisplatin-Induced Nephrotoxicity: Test of Five Protocols of Hydration during Cisplatin Therapy in Rat Models. J Toxicol 2021; 2021:5547341. [PMID: 34646321 PMCID: PMC8505073 DOI: 10.1155/2021/5547341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/01/2021] [Accepted: 09/20/2021] [Indexed: 11/30/2022] Open
Abstract
Backgrounds Cisplatin (CP) still is a novel choice for solid tumor therapy, but it is accompanied with the side effect of nephrotoxicity. Hydration may reduce the risk of CP-induced nephrotoxicity, while the issue is still challenging. In this study, five types of hydration protocols including saline, mannitol, dextrose saline, saline plus furosemide, and saline plus mannitol were examined in both sexes of rats during CP therapy. Methods Seventy-six male and female Wistar rats in 14 groups of experiments were subjected to CP therapy, and five types of hydration protocols were implemented, and the induced nephrotoxicity was evaluated via biochemical markers, kidney function parameters, and pathology investigation. Results Male and female rats had different responses to hydration protocol types. The higher mortality rate was seen in female rats that received mannitol or dextrose hydration types. In addition, the serum levels of blood urea nitrogen (BUN) and creatinine (Cr) and sodium excretion fraction (ENa%) increased and the clearance of Cr (ClCr) decreased significantly (P < 0.05) in female rats hydrated with saline plus furosemide or mannitol plus saline-treated groups. The worsened condition in male rats is observed in the mannitol hydration group with a significant decrease of ClCr and significant increase of serum BUN and Cr and ENa% (P < 0.05). The higher kidney tissue damage score (KTDS) in the mentioned groups verified the findings. Conclusion Hydration with mannitol or dextrose promotes the risk of nephrotoxicity during CP therapy with more intensity on the female.
Collapse
|
373
|
Dong T, Zhang X, Liu Y, Xu S, Chang H, Chen F, Pan L, Hu S, Wang M, Lu M. Opa1 Prevents Apoptosis and Cisplatin-Induced Ototoxicity in Murine Cochleae. Front Cell Dev Biol 2021; 9:744838. [PMID: 34621753 PMCID: PMC8490775 DOI: 10.3389/fcell.2021.744838] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/30/2021] [Indexed: 01/25/2023] Open
Abstract
Optic atrophy1 (OPA1) is crucial for inner mitochondrial membrane (IMM) fusion and essential for maintaining crista structure and mitochondrial morphology. Optic atrophy and hearing impairment are the most prevalent clinical features associated with mutations in the OPA1 gene, but the function of OPA1 in hearing is still unknown. In this study, we examined the ability of Opa1 to protect against cisplatin-induced cochlear cell death in vitro and in vivo. Our results revealed that knockdown of Opa1 affects mitochondrial function in HEI-OC1 and Neuro 2a cells, as evidenced by an elevated reactive oxygen species (ROS) level and reduced mitochondrial membrane potential. The dysfunctional mitochondria release cytochrome c, which triggers apoptosis. Opa1 expression was found to be significantly reduced after cell exposed to cisplatin in HEI-OC1 and Neuro 2a cells. Loss of Opa1 aggravated the apoptosis and mitochondrial dysfunction induced by cisplatin treatment, whereas overexpression of Opa1 alleviated cisplatin-induced cochlear cell death in vitro and in explant. Our results demonstrate that overexpression of Opa1 prevented cisplatin-induced ototoxicity, suggesting that Opa1 may play a vital role in ototoxicity and/or mitochondria-associated cochlear damage.
Collapse
Affiliation(s)
- Tingting Dong
- Biobank of Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuejie Zhang
- Biobank of Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiqing Liu
- Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shan Xu
- Shanghai Ninth People's Hospital, Shanghai Institute of Precision Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haishuang Chang
- Shanghai Ninth People's Hospital, Shanghai Institute of Precision Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengqiu Chen
- Biobank of Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lulu Pan
- Biobank of Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shaoru Hu
- Biobank of Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Wang
- Biobank of Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Lu
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Department of Orthopaedics, Ruijin Hospital, Shanghai Institute of Traumatology and Orthopaedics, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
374
|
Ge X, Jiang Y, Hu X, Yu X. MicroRNA-106a-5p alleviated the resistance of cisplatin in lung cancer cells by targeting Jumonji domain containing 6. Transpl Immunol 2021; 69:101478. [PMID: 34607003 DOI: 10.1016/j.trim.2021.101478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/29/2021] [Accepted: 09/29/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cisplatin (DDP) is used for lung cancer therapy. MicroRNAs, small non-coding RNAs, may contribute to tumorigenesis as well as to drug resistance. We examined regulatory functions of miR-106a-5p in DDP-resistant lung cancer cells. METHODS Differentially expressed miRNAs were provided by Gene Expression Omnibus (GEO) datasets and RT-qPCR examined RNA levels of miR-106a-5p and Jumonji domain-containing protein 6 (JMJD6), an enzyme causing lysine hydroxylation and arginine demethylation. Bindings were determined by luciferase reporter assay. Additionally, the half maximal inhibitory concentration (IC50) of DDP was determined through Cell Counting Kit-8 (CCK-8) after treated by DDP (0, 6.25, 12.5, 25, 50 and 75 μM) and apoptosis rates were analyzed using flow cytometry. Besides that, migratory ability and invasiveness were examined by transwell. Western blot was for measuring protein levels of Bcl-2, Bax in apoptosis and E-cadherin, N-cadherin in epithelial-mesenchymal transition (EMT). RESULTS The IC50 value of DDP-resistant A549 (A549/DDP) cells was higher, so were migration, invasion and N-cadherin in EMT while the apoptosis and E-cadherin in EMT were lower versus the parental A549 cells (no DDP resistance). MiR-106a-5p was low expressed in A549/DDP cells while its overexpression caused decreased migration, invasiveness and EMT but promoted apoptosis. JMJD6 was directly targeted and negatively regulated by miR-106a-5p. Inhibited JMJD6 decreased migratory ability, invasion and EMT but improved apoptosis. Moreover, knockdown of miR-106a-5p induced high level of JMJD6, migration, invasiveness and EMT but low apoptosis rates, which were restrained by JMJD6 suppression. CONCLUSION MiR-106a-5p/JMJD6 axis accelerated cell apoptosis and suppressed invasiveness, migration and EMT in A549/DDP cells.
Collapse
Affiliation(s)
- Xiang Ge
- The Second Affiliated Hospital of Jiaxing University, Pulmonary and Critical Care Medicine, No. 1518, north Huancheng Road Nanhu District, Jiaxing, Zhejiang Province 314000, PR China
| | - Yifei Jiang
- The Second Affiliated Hospital of Jiaxing University, Pulmonary and Critical Care Medicine, No. 1518, north Huancheng Road Nanhu District, Jiaxing, Zhejiang Province 314000, PR China
| | - Xun Hu
- The Second Affiliated Hospital of Jiaxing University, Pulmonary and Critical Care Medicine, No. 1518, north Huancheng Road Nanhu District, Jiaxing, Zhejiang Province 314000, PR China
| | - Xiaoyan Yu
- The Second Affiliated Hospital of Jiaxing University, Pulmonary and Critical Care Medicine, No. 1518, north Huancheng Road Nanhu District, Jiaxing, Zhejiang Province 314000, PR China.
| |
Collapse
|
375
|
Petronijević J, Joksimović N, Milović E, Crnogorac MĐ, Petrović N, Stanojković T, Milivojević D, Janković N. Antitumor activity, DNA and BSA interactions of novel copper(II) complexes with 3,4-dihydro-2(1H)-quinoxalinones. Chem Biol Interact 2021; 348:109647. [PMID: 34520752 DOI: 10.1016/j.cbi.2021.109647] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/16/2021] [Accepted: 09/06/2021] [Indexed: 11/30/2022]
Abstract
In order to discover new therapeutically active agents a series of novel copper(II) complexes with 3,4-dihydro-2(1H)-quinoxalinones were synthesized. All complexes were characterized by IR and EPR spectroscopic techniques and examined for their cytotoxic effect on human cancer cell lines HeLa, LS174, A549 and normal fibroblasts (MRC-5). For further examination of the cytotoxic mechanisms of novel complexes, three of them were chosen for analysing their effects on the distribution of HeLa cells in the cell cycle phases. The results of the flow cytometry analysis suggest that tested complexes lead to time-dependent accumulation of the cells in S and G2/M phases. The strongest accumulation effect showed complex 2d after 48 h of incubation. Competitive experiments with ethidium bromide (EB) indicated that tested compound 2d have affinity to displace EB from the EB-DNA complex through intercalation. Also, the binding parameters values for 2d-BSA complex showed that a reversible 2d-BSA complex is formed and ligand 2d can be stored and carried by BSA.
Collapse
Affiliation(s)
- Jelena Petronijević
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovica 12, Kragujevac, Serbia.
| | - Nenad Joksimović
- University of Kragujevac, Faculty of Science, Department of Chemistry, Radoja Domanovica 12, Kragujevac, Serbia
| | - Emilija Milović
- University of Kragujevac, Institute for Information Technologies Kragujevac, Department of Sciences, Jovana Cvijića bb, 34000, Kragujevac, Serbia
| | | | - Nina Petrović
- Laboratory for Radiobiology and Molecular Genetics, Department of Health and Environment, "VINČA" Institute of Nuclear Sciences -National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Serbia
| | - Tatjana Stanojković
- Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000, Belgrade, Serbia
| | - Dušan Milivojević
- Laboratory for Radiobiology and Molecular Genetics, Department of Health and Environment, "VINČA" Institute of Nuclear Sciences -National Institute of the Republic of Serbia, University of Belgrade, Mike Petrovica Alasa 12-14, Serbia
| | - Nenad Janković
- University of Kragujevac, Institute for Information Technologies Kragujevac, Department of Sciences, Jovana Cvijića bb, 34000, Kragujevac, Serbia
| |
Collapse
|
376
|
Abo El-Magd NF, Ebrahim HA, El-Sherbiny M, Eisa NH. Quinacrine Ameliorates Cisplatin-Induced Renal Toxicity via Modulation of Sirtuin-1 Pathway. Int J Mol Sci 2021; 22:ijms221910660. [PMID: 34639002 PMCID: PMC8508772 DOI: 10.3390/ijms221910660] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 01/05/2023] Open
Abstract
Renal toxicity is a serious side effect that hinders the use of cisplatin, a commonly used and effective chemotherapeutic agent. Meanwhile, quinacrine is an FDA approved drug that has been stated for its anti-inflammatory effect. Thus, we investigated the ameliorative effect of quinacrine against cisplatin-induced renal toxicity. Single intraperitoneal (i.p.) 10 mg/kg cisplatin administration induced renal injury in rats. Our results showed that 10 mg/kg/day quinacrine decreased the mortality rate of rats from 46.15% (cisplatin group) to 12.5%, and significantly decreased renal tissue fibrosis, relative kidney to body weight ratio, serum creatinine and urea levels compared with the cisplatin group. Indeed, quinacrine significantly decreased renal malondialdehyde concentration and increased renal total antioxidant capacity, compared with the cisplatin group. Furthermore, quinacrine caused significant upregulation of renal sirtuin-1 (SIRT-1) with significant downregulation of intercellular adhesion molecule-1 (ICAM-1) and tumor necrosis factor-α (TNF-α). Moreover, quinacrine significantly blocked cisplatin-induced apoptosis, which was made evident by downregulating renal apoptotic proteins (BAX and p53) and upregulating the renal anti-apoptotic protein BCL2, compared with the cisplatin group. In conclusion, this study demonstrates, for the first time, that quinacrine alleviates cisplatin-induced renal toxicity via upregulating SIRT-1, downregulating inflammatory markers (ICAM-1 and TNF-α), reducing oxidative stress, and inhibiting apoptosis.
Collapse
Affiliation(s)
- Nada F. Abo El-Magd
- Biochemistry Department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
- Correspondence: (N.F.A.E.-M.); (N.H.E.)
| | - Hasnaa Ali Ebrahim
- Department of Basic Medical Sciences, College of Medicine, Princess Nourah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia;
- Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 13713, Saudi Arabia;
| | - Nada H. Eisa
- Biochemistry Department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
- Correspondence: (N.F.A.E.-M.); (N.H.E.)
| |
Collapse
|
377
|
Yuan J, Liang X, Zhou W, Feng J, Wang Z, Shen S, Guan X, Zhao L, Deng F. TRPA1 promotes cisplatin-induced nephrotoxicity through inflammation mediated by the MAPK/NF-κB signaling pathway. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1578. [PMID: 34790784 PMCID: PMC8576655 DOI: 10.21037/atm-21-5125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/21/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND The nephrotoxicity induced by cisplatin (DDP) has been a severe obstacle for its clinical use in anticancer treatment. The apoptosis and inflammation induced by DDP are the main causes of the nephrotoxicity. Transient receptor potential ankyrin 1 (TRPA1) is a non-selective cation ligand-gated channel that is involved in the inflammation progress. METHODS The apoptosis, inflammation, MAPK/NF-κB signaling pathway, and TRPA1 expression were assessed after HEK293 cells had been induced by DDP, and the role of TRPA1 in apoptosis and inflammation of DDP-induced HEK293 cells treated with TRPA1 antagonist HC-030031 was also evaluated by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), flow cytometry, and western blot assays. RESULTS The cell viability was reduced by DDP in both a time-dependent and dose-dependent manner with a minimal cytotoxic concentration of 10 μM. Moreover, DDP induced an enhancement of the apoptosis and inflammation in a dose-dependent manner, as indicated by the increase of the relative protein level of cleaved-caspase3 (cleaved-cas3), the cleavage product of caspase-3 substrate poly-ADP-ribose polymerase (cleaved-PARP) and inducible nitric oxide synthase (iNOS), and the messenger RNA (mRNA) expression level of interleukin (IL)-1β, IL-6, tumor necrosis factor-α (TNF-α), and interferon-γ (INF-γ). Additionally, DDP treatment increased the protein phosphorylation expression of IKKβ, JNK, ERK, and p38 in a dose-dependent manner, which was antagonized by the treatment of NF-κB-specific inhibitor BAY 11-7082 and pan-MAPK inhibitor U0126. It was also found that DDP upregulated the expression of TRPA1 at both the mRNA and protein levels in a dose-dependent manner. Besides, block of TRPA1 with HC-030031 relieved the apoptosis, diminished the level of IL-1β, IL-6, TNF-α, and INF-γ, reduced the level of cleaved-cas3, cleaved-PARP, and iNOS, decreased the p-IKKβ, p-JNK, p-ERK, and p-p38 expression, and enhanced the expression of IκBα. CONCLUSIONS Taken together, these results indicate that TRPA1 regulates DDP-induced nephrotoxicity via inflammation mediated by the MAPK/NF-κB signaling pathway in HEK293 cells.
Collapse
Affiliation(s)
- Jinyan Yuan
- Department of Nephrology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao Liang
- Department of Internal Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Wei Zhou
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jing Feng
- Department of Traditional Chinese Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhenyang Wang
- Department of Nephrology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shaoxian Shen
- Department of Nephrology, Jinniu Hospital of Sichuan Provincial People’s Hospital and Chengdu Jinniu District People’s Hospital, Chengdu, China
| | - Xin Guan
- Department of Nephrology, Jinniu Hospital of Sichuan Provincial People’s Hospital and Chengdu Jinniu District People’s Hospital, Chengdu, China
| | - Liangbin Zhao
- Department of Nephrology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fei Deng
- Department of Nephrology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Department of Nephrology, Jinniu Hospital of Sichuan Provincial People’s Hospital and Chengdu Jinniu District People’s Hospital, Chengdu, China
| |
Collapse
|
378
|
Gwon MG, Gu H, Leem J, Park KK. Protective Effects of 6-Shogaol, an Active Compound of Ginger, in a Murine Model of Cisplatin-Induced Acute Kidney Injury. Molecules 2021; 26:5931. [PMID: 34641472 PMCID: PMC8512008 DOI: 10.3390/molecules26195931] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 09/25/2021] [Accepted: 09/28/2021] [Indexed: 12/27/2022] Open
Abstract
Acute kidney injury (AKI) is a dose-limiting side effect of cisplatin therapy in cancer patients. However, effective therapies for cisplatin-induced AKI are not available. Oxidative stress, tubular cell death, and inflammation are known to be the major pathological processes of the disease. 6-Shogaol is a major component of ginger and exhibits anti-oxidative and anti-inflammatory effects. Accumulating evidence suggest that 6-shogaol may serve as a potential therapeutic agent for various inflammatory diseases. However, whether 6-shogaol exerts a protective effect on cisplatin-induced renal side effect has not yet been determined. The aim of this study was to evaluate the effect of 6-shogaol on cisplatin-induced AKI and to investigate its underlying mechanisms. An administration of 6-shogaol after cisplatin treatment ameliorated renal dysfunction and tubular injury, as shown by a reduction in serum levels of creatinine and blood urea nitrogen and an improvement in histological abnormalities. Mechanistically, 6-shogaol attenuated cisplatin-induced oxidative stress and modulated the renal expression of prooxidant and antioxidant enzymes. Apoptosis and necroptosis induced by cisplatin were also suppressed by 6-shogaol. Moreover, 6-shogaol inhibited cisplatin-induced cytokine production and immune cell infiltration. These results suggest that 6-shogaol exhibits therapeutic effects against cisplatin-induced AKI via the suppression of oxidative stress, tubular cell death, and inflammation.
Collapse
Affiliation(s)
- Mi-Gyeong Gwon
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea; (M.-G.G.); (H.G.); (K.-K.P.)
| | - Hyemin Gu
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea; (M.-G.G.); (H.G.); (K.-K.P.)
| | - Jaechan Leem
- Department of Immunology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea
| | - Kwan-Kyu Park
- Department of Pathology, School of Medicine, Catholic University of Daegu, Daegu 42472, Korea; (M.-G.G.); (H.G.); (K.-K.P.)
| |
Collapse
|
379
|
Selective depletion of a CD64-expressing phagocyte subset mediates protection against toxic kidney injury and failure. Proc Natl Acad Sci U S A 2021; 118:2022311118. [PMID: 34518373 PMCID: PMC8488624 DOI: 10.1073/pnas.2022311118] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 01/16/2023] Open
Abstract
Dendritic cells (DC), macrophages, and monocytes, collectively known as mononuclear phagocytes (MPs), critically control tissue homeostasis and immune defense. However, there is a paucity of models allowing to selectively manipulate subsets of these cells in specific tissues. The steady-state adult kidney contains four MP subsets with Clec9a-expression history that include the main conventional DC1 (cDC1) and cDC2 subtypes as well as two subsets marked by CD64 but varying levels of F4/80. How each of these MP subsets contributes to the different phases of acute kidney injury and repair is unknown. We created a mouse model with a Cre-inducible lox-STOP-lox-diphtheria toxin receptor cassette under control of the endogenous CD64 locus that allows for diphtheria toxin-mediated depletion of CD64-expressing MPs without affecting cDC1, cDC2, or other leukocytes in the kidney. Combined with specific depletion of cDC1 and cDC2, we revisited the role of MPs in cisplatin-induced kidney injury. We found that the intrinsic potency reported for CD11c+ cells to limit cisplatin toxicity is specifically attributed to CD64+ MPs, while cDC1 and cDC2 were dispensable. Thus, we report a mouse model allowing for selective depletion of a specific subset of renal MPs. Our findings in cisplatin-induced injury underscore the value of dissecting the functions of individual MP subsets in kidney disease, which may enable therapeutic targeting of specific immune components in the absence of general immunosuppression.
Collapse
|
380
|
Yang Y, Zhu X, Yu G, Ma J. Protective Effect of Pyxinol, One Active Ingredient of Lichenes on Cisplatin-Induced Nephrotoxicity via Ameliorating DNA Damage Response. Front Pharmacol 2021; 12:735731. [PMID: 34552492 PMCID: PMC8450395 DOI: 10.3389/fphar.2021.735731] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 08/23/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Cisplatin is a valuable chemotherapeutic agent against malignant tumors. However, the clinical use of cisplatin is limited by its side effects such as renal injury. Pyxinol is an active constituent of Lichenes and its effects on cisplatin-induced nephrotoxicity is currently unknown. This study aims to examine the potential protective effects of pyxinol on cisplatin-induced renal injury and explore the underlying mechanisms. Methods:In vivo rat model of cisplatin-induced nephrotoxicity was induced by intraperitoneal (i.p) administration of cisplatin. The blood urea nitrogen and creatinine levels were measured and renal histological analysis was conducted to evaluate the renal function; The TUNEL staining, western blotting and real-time PCR assays were conducted to examine related molecular changes. Finally, the in vivo anti-tumor efficacy was examined in the xenograft tumor model using nude mice. Results: Pretreatment with pyxinol attenuated cisplatin-induced increase in blood urea nitrogen, creatinine and urinary protein excretion and the magnitude of injury in the renal tubules. Pyxinol ameliorated the activation of p53 via attenuating the DNA damage response, which then attenuated the tubular cell apoptosis. Finally, pyxinol could potentiate the in vivo anti-tumor efficacy of cisplatin against the xenograft tumor of cervical cancer cells in nude mice. Conclusions: Combining pyxinol with cisplatin could alleviate cisplatin-induced renal injury without decreasing its therapeutic efficacy, which might represent a beneficial adjunct therapy for cisplatin-based chemotherapeutic regimens in the clinic.
Collapse
Affiliation(s)
- Yanting Yang
- Department of Clinical Medicine, Binzhou Medical University, Yantai, China
| | - Xiuhong Zhu
- People's Hospital of Jimo District, Qingdao, China
| | - Guohua Yu
- Department of Clinical Medicine, Binzhou Medical University, Yantai, China.,Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Medical College of Qingdao University, Yantai, China
| | - Jinbo Ma
- Department of Clinical Medicine, Binzhou Medical University, Yantai, China
| |
Collapse
|
381
|
Badar A, Ahmed A, Al-Tamimi DM, Isab AA, Altaf M, Ahmed S. Histological Changes in Renal, Hepatic and Cardiac Tissues of Wistar Rats after 6 Weeks Treatment with Bipyridine Gold (III) Complex with Dithiocarbamate Ligands. Pharmaceutics 2021; 13:pharmaceutics13101530. [PMID: 34683832 PMCID: PMC8539664 DOI: 10.3390/pharmaceutics13101530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/18/2021] [Accepted: 09/20/2021] [Indexed: 11/16/2022] Open
Abstract
Bipyridine gold (III) dithiocarbamate compounds are Gold-III complexes with promising cytotoxic properties. In this study, the subacute toxicity of a Gold (III) complex with dithiocarbamate ligand was evaluated. In the acute toxicity component, an initial LD50 (38.46 mg/kg) was calculated by the administration of 50, 100, 200, 400, and 800 mg/kg of the compound to five groups of rats, respectively (n = 4 each). The sixth group was the control. The sub-acute toxicity component comprised the control group A (n = 6) and the study groups B (n = 10) and C (n = 4), which were administered 1 mL distilled water, 1/10 LD50 (3.8 mg/kg), and 1/5 LD50 (7.6 mg/kg), respectively, daily for 6 weeks. The alive animals were then sacrificed. Autopsy; preservation of renal, hepatic and cardiac tissue in buffered formalin; histopathological processing; microscopic evaluation; and comparison with the controls were sequentially conducted. In the subacute toxicity study at dosages of 3.8 mg/kg and 7.6 mg/kg, the renal tubules remained unaffected with no necrosis or vacuolization. Mild to moderate renal interstitial, hepatic capsular, lobular and portal inflammation along with mild focal hepatic vacuolization were present. At 3.8 mg/kg, the cardiac muscle fibers were unremarkable in 80% (n = 8) of the specimens, with mild focal hyalinization in 20% (n = 2) of the specimens. The same was observed in 50% (n = 2) of the specimens at 7.6 mg/kg. Variable congestion was evident in all of the groups. In the subacute toxicity study, the absence of renal tubular necrosis or vacuolization, the presence of mild inflammatory hepatic and renal alterations, and predominantly unremarkable cardiac muscle fibers suggest that Bipyridine gold (III)-dithiocarbamate is safe in animal studies and is a potential candidate for clinical trials.
Collapse
Affiliation(s)
- Ahmed Badar
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
- Correspondence:
| | - Ayesha Ahmed
- Department of Pathology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia; (A.A.); (D.M.A.-T.)
| | - Dalal M. Al-Tamimi
- Department of Pathology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia; (A.A.); (D.M.A.-T.)
| | - Anvarhusein A. Isab
- Department of Chemistry, King Fahd University of Petroleum and Minerals, Dhahran 31261, Saudi Arabia;
| | - Muhammad Altaf
- Department of Chemistry, Government College University, Lahore 54000, Pakistan;
| | - Sania Ahmed
- Army Medical College, Abid Majeed Road, Rawalpindi 46000, Pakistan;
| |
Collapse
|
382
|
Bai Y, Kim JY, Bisunke B, Jayne LA, Silvaroli JA, Balzer MS, Gandhi M, Huang KM, Sander V, Prosek J, Cianciolo RE, Baker SD, Sparreboom A, Jhaveri KD, Susztak K, Bajwa A, Pabla NS. Kidney toxicity of the BRAF-kinase inhibitor vemurafenib is driven by off-target ferrochelatase inhibition. Kidney Int 2021; 100:1214-1226. [PMID: 34534550 DOI: 10.1016/j.kint.2021.08.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 07/21/2021] [Accepted: 08/13/2021] [Indexed: 12/29/2022]
Abstract
A multitude of disease and therapy related factors drive the frequent development of kidney disorders in cancer patients. Along with chemotherapy, the newer targeted therapeutics can also cause kidney dysfunction through on and off-target mechanisms. Interestingly, among the small molecule inhibitors approved for the treatment of cancers that harbor BRAF-kinase activating mutations, vemurafenib can trigger tubular damage and acute kidney injury. BRAF is a proto-oncogene involved in cell growth. To investigate the underlying mechanisms, we developed cell culture and mouse models of vemurafenib kidney toxicity. At clinically relevant concentrations vemurafenib induces cell-death in transformed and primary mouse and human kidney tubular epithelial cells. In mice, two weeks of daily vemurafenib treatment causes moderate acute kidney injury with histopathological characteristics of kidney tubular epithelial cells injury. Importantly, kidney tubular epithelial cell-specific BRAF gene deletion did not influence kidney function under normal conditions or alter the severity of vemurafenib-associated kidney impairment. Instead, we found that inhibition of ferrochelatase, an enzyme involved in heme biosynthesis contributes to vemurafenib kidney toxicity. Ferrochelatase overexpression protected kidney tubular epithelial cells and conversely ferrochelatase knockdown increased the sensitivity to vemurafenib-induced kidney toxicity. Thus, our studies suggest that vemurafenib-associated kidney tubular epithelial cell dysfunction and kidney toxicity is BRAF-independent and caused, in part, by off-target ferrochelatase inhibition.
Collapse
Affiliation(s)
- Yuntao Bai
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Ji Young Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Bijay Bisunke
- Department of Genetics, Genomics, and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Laura A Jayne
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Josie A Silvaroli
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Michael S Balzer
- Department of Medicine and Genetics, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Megha Gandhi
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Kevin M Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Veronika Sander
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Jason Prosek
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Rachel E Cianciolo
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Sharyn D Baker
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA
| | - Kenar D Jhaveri
- Division of Kidney Diseases and Hypertension, Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Northwell Health, Great Neck, New York, USA
| | - Katalin Susztak
- Department of Medicine and Genetics, Renal Electrolyte and Hypertension Division, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Amandeep Bajwa
- Department of Genetics, Genomics, and Informatics, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA; Department of Microbiology, Immunology, and Biochemistry, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA; Transplant Research Institute, James D. Eason Transplant Institute, Department of Surgery, College of Medicine, The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Navjot Singh Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, USA.
| |
Collapse
|
383
|
Kim MH, Yuk HD, Jeong CW, Kwak C, Kim HH, Ku JH. Estimated Glomerular Filtration Rate as a Prognostic Factor in Urothelial Carcinoma of the Upper Urinary Tract: A Systematic Review and Meta-Analysis. J Clin Med 2021; 10:4155. [PMID: 34575266 PMCID: PMC8472261 DOI: 10.3390/jcm10184155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/03/2021] [Accepted: 09/14/2021] [Indexed: 11/18/2022] Open
Abstract
Preoperative renal function is associated with worse outcomes in patients undergoing radical nephroureterectomy (RNU). The purpose of this systemic review and meta-analysis was to determine the association of preoperative renal function with oncological outcome in patients who underwent RNU. We searched articles published up to March 2021 in PubMed, Scopus, and Embase by combining "urothelial carcinoma", "radical nephroureterectomy", and "estimated glomerular filtration rate". We also manually screened the reference list for publications following general guidelines recommended by the preferred reporting items for systematic reviews and meta-analyses (PRISMA) statement. The relationship between preoperative renal function and survival was expressed as overall survival (OS), progression-free survival (PFS), and cancer-specific survival (CSS). This review and meta-analysis included 13 studies involving a total of 4668 patients who received RNU. Pooled analysis showed significant negative association of preoperative renal function with PFS (HR: 1.51, 95% CI: 1.23-1.80, p < 0.00001), CSS (HR: 1.63, 95% CI: 1.38-1.92, p < 0.00001), and OS (HR: 1.22, 95% CI: 1.10-1.35, p < 0.00001). Patients with upper tract urothelial carcinoma (UTUC) who received RNU showed a significant negative association of preoperative renal function with survival.
Collapse
Affiliation(s)
| | | | | | | | | | - Ja Hyeon Ku
- Department of Urology, Seoul National University College of Medicine, Seoul National University Hospital, Seoul 03080, Korea; (M.H.K.); (H.D.Y.); (C.W.J.); (C.K.); (H.H.K.)
| |
Collapse
|
384
|
Ashry M, Galal El-Sahra D, Gaber DA, A Mustafa M, Abdel-Wahhab KG. Nephroprotective Effect of Costus ( Saussurea costus) Ethanolic Extract on Oxaliplatin ®-induced Nephrotoxicity in Adult Male Wistar Rats. Pak J Biol Sci 2021; 24:830-839. [PMID: 34486350 DOI: 10.3923/pjbs.2021.830.839] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
<b>Background and Objective:</b> Oxaliplatin<sup>®</sup> is an antineoplastic platinum-based compound; nephrotoxicity is one of its most serious side effects. This study aimed to explore the nephroprotective potential of Costus Ethanolic Extract (CEE) against Oxaliplatin<sup>®</sup>-induced nephrotoxicity. <b>Materials and Methods:</b> Adult male Wistar rats, weighting 140-160 g, were randomly divided into four groups: (1) Normal rats, (2) Rats ingested with CEE (67.08 mg kg<sup>1</sup> day<sup>1</sup>), (3) Rats injected (ip) with Oxaliplatin<sup>®</sup> (10 mg kg<sup>1</sup> week<sup>1</sup>) and (4) rats treated with CEE in combination Oxaliplatin<sup>®</sup> injection. <b>Results:</b> After six weeks of treatments, the results revealed that CEE ingestion along with Oxaliplatin<sup>®</sup> injection markedly minimized the Oxaliplatin<sup>®</sup>-induced renal deterioration; this was evidenced by the significant reduction in serum urea, creatinine, uric acid, Tumor Necrosis Factor Alpha (TNF-α), Interleukin 1Beta (IL<sup>1</sup>β) and Sodium ion (Na<sup>+</sup>) levels as well as kidney Malondialdehyde (MDA), Nitric Oxide (NO) and DNA fragmentation values. Controversially, a marked rise in serum Calcium, Potassium Ion (K<sup>+</sup>) and Cluster of Differentiation 4 (CD4) levels besides renal Glutathione (GSH), Catalase (CAT) and Superoxide Dismutase (SOD) values. Similarly, the histopathological findings confirmed the biochemical ones as the CEE restored the Oxaliplatin<sup>®</sup>-induced histological degenerations. <b>Conclusion:</b> In conclusion, CEE exhibited nephron-protection efficiency against Oxaliplatin<sup>®</sup>-induced nephrotoxicity; this promising effect may be achieved through the antioxidant and radical scavenging activities of its constituents.
Collapse
|
385
|
El-Far YM, El-Mesery M. Pevonedistat attenuates cisplatin-induced nephrotoxicity in mice by downregulating the release of inflammatory mediators. J Biochem Mol Toxicol 2021; 35:e22908. [PMID: 34476871 DOI: 10.1002/jbt.22908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 07/09/2021] [Accepted: 08/20/2021] [Indexed: 01/11/2023]
Abstract
Pevonedistat (MLN4924) is a specific NEDD8-activating enzyme inhibitor that inactivates cullin-RING ligases involved in ubiquitylation and turnover of different signaling molecules. In the current study, we evaluated the effect of pevonedistat on cisplatin (CIS)-induced nephrotoxicity in mice. Serum creatinine and urea levels were analyzed in different groups. Histopathological examination of renal tissue was done using hematoxylin and eosin staining. In addition, renal IL-6 and TNF-α expressions were analyzed using the enzyme-linked immunosorbent assay technique, and IL-1β and NF-κB expressions were analyzed by immunohistochemical staining of renal tissue. Caspase-3, A20, β-catenin, and Nrf2 gene expressions in renal tissue were analyzed using the reverse-transcription polymerase chain reaction technique. Western blot analysis was adopted to assess cleaved caspase-3 and β-catenin expressions in renal tissue. Pevonedistat coadministration with CIS improved kidney functions and attenuated CIS-induced nephrotoxicity as indicated by the significant decrease in serum creatinine and urea levels. In addition, pevonedistat coadministration with CIS showed a significant decrease in caspase-3 and a significant increase in A20, β-catenin, and Nrf2 gene expressions. Also, pevonedistat decreased caspase-3 cleavage to p19 in mice treated with CIS. Moreover, pevonedistat decreased CIS-induced upregulation of IL-6, TNF-α, IL-1β, and NF-κB protein expressions in renal tissue. Thus, pevonedistat alleviated CIS-induced nephrotoxicity that might be attributed to suppression of the inflammation induced in renal tissue.
Collapse
Affiliation(s)
- Yousra M El-Far
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Mesery
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
386
|
Ehsan N, Ijaz MU, Ashraf A, Sarwar S, Samad A, Afzal G, Andleeb R, Al-Misned FA, Al-Ghanim KA, Ahmed Z, Riaz MN, Mahboob S. Mitigation of cisplatin induced nephrotoxicity by casticin in male albino rats. BRAZ J BIOL 2021; 83:e243438. [PMID: 34468509 DOI: 10.1590/1519-6984.243438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/08/2021] [Indexed: 11/22/2022] Open
Abstract
Cisplatin (CP) is a commonly used, powerful antineoplastic drug, having numerous side effects. Casticin (CAS) is considered as a free radical scavenger and a potent antioxidant. The present research was planned to assess the curative potential of CAS on CP persuaded renal injury in male albino rats. Twenty four male albino rats were distributed into four equal groups. Group-1 was considered as a control group. Animals of Group-2 were injected with 5mg/kg of CP intraperitoneally. Group-3 was co-treated with CAS (50mg/kg) orally and injection of CP (5mg/kg). Group-4 was treated with CAS (50mg/kg) orally throughout the experiment. CP administration substantially reduced the activities of catalase (CAT), superoxide dismutase (SOD), peroxidase (POD), glutathione S-transferase (GST), glutathione reductase (GSR), glutathione (GSH) content while increased thiobarbituric acid reactive substances (TBARS), and hydrogen peroxide (H2O2) levels. Urea, urinary creatinine, urobilinogen, urinary proteins, kidney injury molecule-1 (KIM-1), and neutrophil gelatinase-associated lipocalin (NGAL) levels were substantially increased. In contrast, albumin and creatinine clearance was significantly reduced in CP treated group. The results demonstrated that CP significantly increased the inflammation indicators including nuclear factor kappa-B (NF-κB), tumor necrosis factor-α (TNF-α), Interleukin-1β (IL-1β), Interleukin-6 (IL-6) levels and cyclooxygenase-2 (COX-2) activity and histopathological damages. However, the administration of CAS displayed a palliative effect against CP-generated renal toxicity and recovered all parameters by bringing them to a normal level. These results revealed that the CAS is an effective compound having the curative potential to counter the CP-induced renal damage.
Collapse
Affiliation(s)
- N Ehsan
- University of Agriculture - UAF, Faculty of Science, Department of Zoology, Faisalabad, Pakistan
| | - M U Ijaz
- University of Agriculture - UAF, Faculty of Science, Department of Zoology, Faisalabad, Pakistan
| | - A Ashraf
- Government College University - GCUF, Faculty of Life Science, Department Zoology, Faisalabad, Pakistan
| | - S Sarwar
- University of Agriculture - UAF, Faculty of Science, Department of Zoology, Faisalabad, Pakistan
| | - A Samad
- University of Agriculture - UAF, Faculty of Science, Department of Zoology, Faisalabad, Pakistan
| | - G Afzal
- Islamia University - IUB, Department of Zoology, Bahawalpur, Pakistan
| | - R Andleeb
- Government College University - GCUF, Faculty of Life Science, Department Zoology, Faisalabad, Pakistan
| | - F A Al-Misned
- King Saud University - KSU, College of Science, Department of Zoology, Riyadh, Saudi Arabia
| | - K A Al-Ghanim
- King Saud University - KSU, College of Science, Department of Zoology, Riyadh, Saudi Arabia
| | - Z Ahmed
- King Saud University - KSU, College of Science, Department of Zoology, Riyadh, Saudi Arabia
| | - M N Riaz
- Texas A&M University, AMU, Brazos, Texas, United States of America
| | - S Mahboob
- King Saud University - KSU, College of Science, Department of Zoology, Riyadh, Saudi Arabia
| |
Collapse
|
387
|
Ikeda Y, Hamano H, Horinouchi Y, Miyamoto L, Hirayama T, Nagasawa H, Tamaki T, Tsuchiya K. Role of ferroptosis in cisplatin-induced acute nephrotoxicity in mice. J Trace Elem Med Biol 2021; 67:126798. [PMID: 34087581 DOI: 10.1016/j.jtemb.2021.126798] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/02/2021] [Accepted: 05/25/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Cisplatin is widely used as an antitumor drug for the treatment of solid tumors. However, its use has been limited owing to nephrotoxicity, a major side effect. The mechanism of cisplatin-induced nephrotoxicity (CIN) has long been investigated in order to develop preventive/therapeutic drugs. Ferroptosis is a newly identified form of non-apoptotic regulated cell death induced by iron-mediated lipid peroxidation and is involved in the pathophysiology of various diseases. In this study, we examined the role of ferroptosis in CIN. METHODS We evaluated the role of ferroptosis in CIN by in vivo experiments in a mouse model. RESULTS Cisplatin increased the protein expressions of transferrin receptor-1 and ferritin, and iron content in the kidney of mice. In addition, treatment with cisplatin augmented renal ferrous iron and hydroxyl radical levels with co-localization. Mice administered cisplatin demonstrated kidney injury, with renal dysfunction and increased inflammatory cytokine expression; these changes were ameliorated by Ferrostatin-1 (Fer-1), an inhibitor of ferroptosis. The expression of the ferroptosis markers, COX2 and 4-hydroxynonenal (4-HNE), increased with cisplatin administration, and decreased with the administration of Fer-1. By contrast, cisplatin-induced apoptosis and necroptosis were inhibited by treatment with Fer-1. Moreover, deferoxamine, an iron chelator, also inhibited CIN, with a decrease in the expression of COX-2 and 4-HNE. CONCLUSION Ferroptosis is involved in the pathogenesis of CIN and might be used as a new preventive target for CIN.
Collapse
Affiliation(s)
- Yasumasa Ikeda
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan.
| | - Hirofumi Hamano
- Department of Pharmacy, Tokushima University Hospital, Tokushima, Japan
| | - Yuya Horinouchi
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Licht Miyamoto
- Department of Medical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Tasuku Hirayama
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, Gifu, Japan
| | - Hideko Nagasawa
- Laboratory of Pharmaceutical and Medicinal Chemistry, Gifu Pharmaceutical University, Gifu, Japan
| | - Toshiaki Tamaki
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan; Anan-Medical Center, Anan, Japan
| | - Koichiro Tsuchiya
- Department of Medical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
388
|
Rosner MH, DeMauro Renaghan A. Disorders of Divalent Ions (Magnesium, Calcium, and Phosphorous) in Patients With Cancer. Adv Chronic Kidney Dis 2021; 28:447-459.e1. [PMID: 35190111 DOI: 10.1053/j.ackd.2021.09.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/23/2021] [Accepted: 09/08/2021] [Indexed: 12/15/2022]
Abstract
Disorders of the divalent ions (magnesium, calcium, and phosphorous) are frequently encountered in patients with cancer. Of these, hypomagnesemia, hypocalcemia, hypercalcemia, and hypophosphatemia are seen most commonly. These electrolyte disturbances may be related to the underlying malignancy or due to side effects of anticancer therapy. When caused by a paraneoplastic process, these abnormalities may portend a poor prognosis. Importantly, the development of severe electrolyte derangements may be associated with symptoms that negatively impact quality of life, preclude the administration of critical chemotherapeutic agents, or lead to life-threatening complications that require hospitalization and emergent treatment. In accordance, prompt recognition and treatment of these disorders is key to improving outcomes in patients living with cancer. This review will discuss selected derangements of the divalent ions seen in this population, with a focus on paraneoplastic and therapy-associated etiologies.
Collapse
|
389
|
Gupta S, Portales-Castillo I, Daher A, Kitchlu A. Conventional Chemotherapy Nephrotoxicity. Adv Chronic Kidney Dis 2021; 28:402-414.e1. [PMID: 35190107 DOI: 10.1053/j.ackd.2021.08.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 12/27/2022]
Abstract
Conventional chemotherapies remain the mainstay of treatment for many malignancies. Kidney complications of these therapies are not infrequent and may have serious implications for future kidney function, cancer treatment options, eligibility for clinical trials, and overall survival. Kidney adverse effects may include acute kidney injury (via tubular injury, tubulointerstitial nephritis, glomerular disease and thrombotic microangiopathy), long-term kidney function loss and CKD, and electrolyte disturbances. In this review, we summarize the kidney complications of conventional forms of chemotherapy and, where possible, provide estimates of incidence, and identify risk factors and strategies for kidney risk mitigation. In addition, we provide recommendations regarding kidney dose modifications, recognizing that these adjustments may be limited by available supporting pharmacokinetic and clinical outcomes data. We discuss management strategies for kidney adverse effects associated with these therapies with drug-specific recommendations. We focus on frequently used anticancer agents with established kidney complications, including platinum-based chemotherapies (cisplatin, carboplatin, oxaliplatin), cyclophosphamide, gemcitabine, ifosfamide, methotrexate and pemetrexed, among others.
Collapse
|
390
|
Mechanism Prediction of Astragalus membranaceus against Cisplatin-Induced Kidney Damage by Network Pharmacology and Molecular Docking. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:9516726. [PMID: 34457031 PMCID: PMC8390139 DOI: 10.1155/2021/9516726] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/21/2021] [Accepted: 08/11/2021] [Indexed: 02/05/2023]
Abstract
Background Cisplatin is a frequently used and effective chemotherapy drug in clinical practice, but severe side effects limit its use, among which nephrotoxicity is considered the most serious and prolonged damage to the body. Astragalus membranaceus (AM) is a well-known herbal medicine, and modern pharmacological studies have confirmed its antioxidant, immunomodulatory, and antiapoptotic effects. Clinical studies have shown that AM and its active components can attenuate cisplatin-induced kidney damage, but the molecular mechanism has not been fully expounded. Materials and Methods First, the components and targets information of AM were collected from the TCMSP, and the relevant targets of cisplatin-induced kidney damage were accessed from the GeneCards and OMIM databases. Then, the core targets were selected by the Venn diagram and network topology analysis, which was followed by GO and KEGG pathway enrichment analysis. Finally, we construct a component-target-pathway network. Furthermore, molecular docking was carried out to identify the binding activity between active components and key targets. Results A total of 20 active components and 200 targets of AM and 646 targets related to cisplatin-induced kidney damage were obtained. 91 intersection targets were found between AM and cisplatin-induced kidney damage. Then, 16 core targets were identified, such as MAPK1, TNF-α, and p53. Furthermore, GO and KEGG pathway enrichment analysis suggested that MAPK, Toll-like receptor, and PI3K-Akt signaling pathways may be of significance in the treatment of cisplatin-induced kidney damage by AM. Molecular docking indicated that quercetin and kaempferol had high binding affinities with many core targets. Conclusion In summary, the active components, key targets, and signaling pathways of AM in the treatment of cisplatin-induced kidney damage were predicted in this study, which contributed to the development and application of AM.
Collapse
|
391
|
Zhang JJ, Zhou YD, Liu YB, Wang JQ, Li KK, Gong XJ, Lin XH, Wang YP, Wang Z, Li W. Protective Effect of 20(R)-Ginsenoside Rg3 Against Cisplatin-Induced Renal Toxicity via PI3K/AKT and NF-[Formula: see text]B Signaling Pathways Based on the Premise of Ensuring Anticancer Effect. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1739-1756. [PMID: 34461812 DOI: 10.1142/s0192415x21500828] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Although the protective effect of ginsenoside on cisplatin-induced renal injury has been extensively studied, whether ginsenoside interferes with the antitumor effect of cisplatin has not been confirmed. In this paper, we verified the main molecular mechanism of 20(R)-ginsenoside Rg3 (R-Rg3) antagonizing cisplatin-induced acute kidney injury (AKI) through the combination of in vivo and in vitro models. It is worth mentioning that the two cell models of HK-2 and HepG2 were used simultaneously for the first time to explore the effect of the activation site of tumor-associated protein p53 on apoptosis and tumor suppression. The results showed that a single injection of cisplatin (20 mg/kg) led to weight loss, the kidney index of the mice increased, and creatinine (CRE) and blood urea nitrogen (BUN) levels in mice sharply increased. Continuous administration of R-Rg3 at doses of 10 and 20 mg/kg for 10 days could significantly alleviate this symptom. Similarly, R-Rg3 treatment reduced oxidative stress damage caused by cisplatin. Moreover, R-Rg3 could observably reduce the apoptosis and inflammatory infiltration of renal tubular cells induced by cisplatin. We used western blotting analysis to demonstrate that R-Rg3 restored cisplatin-induced AKI might be related to PI3K/AKT and NF-[Formula: see text]B mediated apoptosis and inflammation pathways. In the meantime, we also verified that R-Rg3 could activate different sites of p53 to control renal cell apoptosis induced by cisplatin without affecting its antitumor effect.
Collapse
Affiliation(s)
- Jun-Jie Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Yan-Dan Zhou
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Yong-Bo Liu
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China
| | - Jian-Qiang Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Ke-Ke Li
- Key Laboratory of Biotechnology and Bioresources Utilization, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| | - Xiao-Jie Gong
- Key Laboratory of Biotechnology and Bioresources Utilization, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| | - Xiang-Hui Lin
- Liaoning XIFENG Pharmaceutical Group Co., Ltd., Huanren 117200, P. R. China
| | - Ying-Ping Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun 130118, P. R. China.,National & Local Joint Engineering Research Center for Ginseng Breeding and Development, Changchun 130118, P. R. China.,Key Laboratory of Biotechnology and Bioresources Utilization, College of Life Science, Dalian Minzu University, Dalian 116600, P. R. China
| |
Collapse
|
392
|
Zhang Y, Zhou Y, Zhang H, Tian L, Hao J, Yuan Y, Li W, Liu Y. DNA binding and evaluation of anticancer activity in vitro and in vivo of iridium(III) polypyridyl complexes. J Inorg Biochem 2021; 224:111580. [PMID: 34438219 DOI: 10.1016/j.jinorgbio.2021.111580] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/01/2021] [Accepted: 08/11/2021] [Indexed: 01/25/2023]
Abstract
In this report, we synthesized three new iridium(III) complexes: [Ir(piq)2(apip)]PF6 (Ir1, piq = 1-phenylisoquinoline, apip = 2-aminophenyl-1H-imidazo[4,5-f][1,10]phenanthroline), [Ir(piq)2(maip)]PF6 (Ir2, maip = 3-aminophenyl-1H-imidazo[4,5-f][1,10]phenanthroline) and [Ir(piq)2(paip)]PF6 (Ir3, paip = 4-aminophenyl-1H-imidazo[4,5-f][1,10]phenanthroline). The DNA binding was investigated. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method was used to detect the cytotoxic activity of Ir1, Ir2 and Ir3, the complexes show highly active against B16 cells with IC50 values of 0.3 ± 0.2 μM, 3.7 ± 0.2 μM and 4.6 ± 1.1 μM, respectively. Subsequently, cellular uptake suggested that the cytotoxicity of the complexes is attributed to their differences in cellular uptake levels. In addition, complexes Ir1, Ir2 and Ir3 induce cell cycle arrest at the G0/G1 phase and regulate the cell cycle mediators such as cyclin D1, CDK6 (cyclin-dependent kinase 6), CDK4 and p21, leading to the inhibition of B16 cells proliferation. The autophagy was investigated by monodansylcadaverine (MDC) staining. The complexes can promote the change from LC3-I to LC3-II, up-regulate levels of Beclin-1 and down-regulate expression of p62. The complexes induced apoptosis by regulating the expression levels of related indicators such as PARP (poly ADP-ribose polymerase), PI3K (phosphoinositide-3 kinase), AKT (protein kinase B), Caspase, Bcl-2 (B-cell lymphoma-2), Bad (Bcl2 associated death promoter), Bax (Bcl2-associated X) and Cyto C (cytochrome C). Additionally, Ir1 exerted significant antitumor activity in the suppression of malignant melanoma proliferation in vivo. As indicated in the above results, these complexes were highly effective for malignant melanoma treatment through the intrinsic pathway and provided much insight into anticancer drugs for tumor therapy.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yi Zhou
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Huiwen Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Li Tian
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Jing Hao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yuhan Yuan
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Wenlong Li
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| |
Collapse
|
393
|
Afsar T, Razak S, Aldisi D, Shabbir M, Almajwal A, Al Kheraif AA, Arshad M. Acacia hydaspica R. Parker ethyl-acetate extract abrogates cisplatin-induced nephrotoxicity by targeting ROS and inflammatory cytokines. Sci Rep 2021; 11:17248. [PMID: 34446789 PMCID: PMC8390681 DOI: 10.1038/s41598-021-96509-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 08/11/2021] [Indexed: 11/16/2022] Open
Abstract
Cisplatin (CisPT) is a chemotherapeutic drug that outcomes in adverse effects. In this study, we examined the effect of A. hydaspica ethyl acetate extract (AHE) in an animal model of cisplatin-induced acute kidney injury (AKI). 36 male Sprague Dawley rats were used in the AKI rat model, and CisPT (7.5 mg/kg BW, i.p) single dose was given. In the pretreatment module, AHE (400 mg/kgBW/day, p.o) was given for 7 days before and after CisPT injection. While in the post-treatment group AHE was administered for 7 days after a single CisPT shot. The standard group received silymarin (100 mg/kg BW, p.o) for 7 days before and after CisPT injection. In HCT 116 tumor xenografts (n = 32) two groups of mice were pretreated with 400 mg/kg AHE orally for 7 days and two groups were treated with distilled water. On day 7 of pretreatment one distilled water and one AHE pretreated group were injected i.p with 15 mg/kg bw dose followed by another dose of CisPT 2 wk later. AHE groups were additionally treated with 400 mg/kg AHE for 3 days/week for 2 weeks. CisPT significantly deteriorated renal function parameters, i.e., PH, specific gravity, total protein, albumin, urea, creatinine, uric acid, globulin and blood urea nitrogen. CisPT treatment increased oxidative stress markers, while lower renal antioxidant enzymes. AHE pretreatment ameliorates significantly (p < 0.0001) CisPT-induced alterations in serum and urine markers for kidney function. Furthermore, AHE pretreatment more efficiently (p < 0.001) decreases oxidative stress markers, attenuate NF-κB, and IL-6 protein and mRNA expression by augmenting antioxidant enzyme levels compared to post-treatment. The histological observations verified the protective effect of AHE. In tumor xenograft mice, AHE treatment significantly reduced CisPT induced oxidative stress while it did not interfere with the anticancer efficacy of cisplatin as shown by significance (p < 0.001) decrease in tumor size after treatment. A. hydaspica AHE might provide a prospective adjuvant that precludes CisPT-induced nephrotoxicity without compromising its antitumor potential.
Collapse
Affiliation(s)
- Tayyaba Afsar
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia.
| | - Suhail Razak
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia.
| | - Dara Aldisi
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Maria Shabbir
- Atta-Ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Ali Almajwal
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Abdulaziz Abdullah Al Kheraif
- Dental Biomaterials Research Chair, Dental Health Department, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Mohammed Arshad
- Dental Biomaterials Research Chair, Dental Health Department, College of Applied Medical Sciences, King Saud University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
394
|
Regression Modeling of the Antioxidant-to-Nephroprotective Relation Shows the Pivotal Role of Oxidative Stress in Cisplatin Nephrotoxicity. Antioxidants (Basel) 2021; 10:antiox10091355. [PMID: 34572987 PMCID: PMC8464812 DOI: 10.3390/antiox10091355] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 12/14/2022] Open
Abstract
The clinical utility of the chemotherapeutic drug cisplatin is significantly limited by its nephrotoxicity, which is characterized by electrolytic disorders, glomerular filtration rate decline, and azotemia. These alterations are consequences of a primary tubulopathy causing injury to proximal and distal epithelial cells, and thus tubular dysfunction. Oxidative stress plays a role in cisplatin nephrotoxicity and cytotoxicity, but its relative contribution to overall toxicity remains unknown. We studied the relation between the degree of oxidative reduction (provided by antioxidant treatment) and the extent of nephrotoxicity amelioration (i.e., nephroprotection) by means of a regression analysis of studies in animal models. Our results indicate that a linear relation exists between these two parameters, and that this relation very nearly crosses the value of maximal nephroprotection at maximal antioxidant effect, suggesting that oxidative stress seems to be a pivotal and mandatory mechanism of cisplatin nephrotoxicity, and, hence, an interesting, rationale-based target for clinical use. Our model also serves to identify antioxidants with enhanced effectiveness by comparing their actual nephroprotective power with that predicted by their antioxidant effect. Among those, this study identified nanoceria, erythropoietin, and maltol as highly effective candidates affording more nephroprotection than expected from their antioxidant effect for prospective clinical development.
Collapse
|
395
|
Adhikari A, Mondal S, Chatterjee T, Das M, Biswas P, Ghosh R, Darbar S, Alessa H, Althakafy JT, Sayqal A, Ahmed SA, Das AK, Bhattacharyya M, Pal SK. Redox nanomedicine ameliorates chronic kidney disease (CKD) by mitochondrial reconditioning in mice. Commun Biol 2021; 4:1013. [PMID: 34446827 PMCID: PMC8390471 DOI: 10.1038/s42003-021-02546-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Accepted: 08/02/2021] [Indexed: 12/29/2022] Open
Abstract
Targeting reactive oxygen species (ROS) while maintaining cellular redox signaling is crucial in the development of redox medicine as the origin of several prevailing diseases including chronic kidney disease (CKD) is linked to ROS imbalance and associated mitochondrial dysfunction. Here, we have shown that a potential nanomedicine comprising of Mn3O4 nanoparticles duly functionalized with biocompatible ligand citrate (C-Mn3O4 NPs) can maintain cellular redox balance in an animal model of oxidative injury. We developed a cisplatin-induced CKD model in C57BL/6j mice with severe mitochondrial dysfunction and oxidative distress leading to the pathogenesis. Four weeks of treatment with C-Mn3O4 NPs restored renal function, preserved normal kidney architecture, ameliorated overexpression of pro-inflammatory cytokines, and arrested glomerulosclerosis and interstitial fibrosis. A detailed study involving human embryonic kidney (HEK 293) cells and isolated mitochondria from experimental animals revealed that the molecular mechanism behind the pharmacological action of the nanomedicine involves protection of structural and functional integrity of mitochondria from oxidative damage, subsequent reduction in intracellular ROS, and maintenance of cellular redox homeostasis. To the best of our knowledge, such studies that efficiently treated a multifaceted disease like CKD using a biocompatible redox nanomedicine are sparse in the literature. Successful clinical translation of this nanomedicine may open a new avenue in redox-mediated therapeutics of several other diseases (e.g., diabetic nephropathy, neurodegeneration, and cardiovascular disease) where oxidative distress plays a central role in pathogenesis.
Collapse
Affiliation(s)
- Aniruddha Adhikari
- Department of Chemical, Biological and Macromolecular Sciences, S. N. Bose National Centre for Basic Sciences, Kolkata, India
| | - Susmita Mondal
- Department of Chemical, Biological and Macromolecular Sciences, S. N. Bose National Centre for Basic Sciences, Kolkata, India
| | | | - Monojit Das
- Department of Zoology, Uluberia College, University of Calcutta, Uluberia, Howrah, India
- Department of Zoology, Vidyasagar University, Rangamati, Midnapore, India
| | - Pritam Biswas
- Department of Microbiology, St. Xavier's College, Kolkata, India
| | - Ria Ghosh
- Department of Biochemistry, University of Calcutta, Kolkata, India
| | - Soumendra Darbar
- Research & Development Division, Dey's Medical Stores (Mfg.) Ltd, Kolkata, India
| | - Hussain Alessa
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Jalal T Althakafy
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ali Sayqal
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Saleh A Ahmed
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
- Chemistry Department, Faculty of Science, Assiut University, Assiut, Egypt
| | - Anjan Kumar Das
- Department of Pathology, Calcutta National Medical College and Hospital, Kolkata, India
| | | | - Samir Kumar Pal
- Department of Chemical, Biological and Macromolecular Sciences, S. N. Bose National Centre for Basic Sciences, Kolkata, India.
- Department of Zoology, Uluberia College, University of Calcutta, Uluberia, Howrah, India.
| |
Collapse
|
396
|
Mapuskar KA, Steinbach EJ, Zaher A, Riley DP, Beardsley RA, Keene JL, Holmlund JT, Anderson CM, Zepeda-Orozco D, Buatti JM, Spitz DR, Allen BG. Mitochondrial Superoxide Dismutase in Cisplatin-Induced Kidney Injury. Antioxidants (Basel) 2021; 10:antiox10091329. [PMID: 34572961 PMCID: PMC8469643 DOI: 10.3390/antiox10091329] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Cisplatin is a chemotherapy agent commonly used to treat a wide variety of cancers. Despite the potential for both severe acute and chronic side effects, it remains a preferred therapeutic option for many malignancies due to its potent anti-tumor activity. Common cisplatin-associated side-effects include acute kidney injury (AKI) and chronic kidney disease (CKD). These renal injuries may cause delays and potentially cessation of cisplatin therapy and have long-term effects on renal function reserve. Thus, developing mechanism-based interventional strategies that minimize cisplatin-associated kidney injury without reducing efficacy would be of great benefit. In addition to its action of cross-linking DNA, cisplatin has been shown to affect mitochondrial metabolism, resulting in mitochondrially derived reactive oxygen species (ROS). Increased ROS formation in renal proximal convoluted tubule cells is associated with cisplatin-induced AKI and CKD. We review the mechanisms by which cisplatin may induce AKI and CKD and discuss the potential of mitochondrial superoxide dismutase mimetics to prevent platinum-associated nephrotoxicity.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Emily J. Steinbach
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Amira Zaher
- Biomedical Science Program, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA;
| | - Dennis P. Riley
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Robert A. Beardsley
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Jeffery L. Keene
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Jon T. Holmlund
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Carryn M. Anderson
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Diana Zepeda-Orozco
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Nephrology, Department of Pediatrics, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - John M. Buatti
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
- Correspondence: ; Tel.: +1-319-335-8019; Fax: +1-319-335-8039
| |
Collapse
|
397
|
Abstract
Magnesium (Mg2+) is the second most abundant intracellular and fourth extracellular cation found in the body and is involved in a wide range of functions in the human cell and human physiology. Its role in most of the enzyme processes (ATP-ases)-stabilisation of nucleic acids (DNA, RNA), regulation of calcium and potassium ion channels, proliferation, glucose metabolism and apoptosis-make it one of the most important cations in the cell. Three pathogenetic mechanisms are mainly implicated in the development of hypomagnesaemia: reduced food intake, decreased intestinal absorption and increased renal excretion of Mg2+. This review presents the function of Mg2+, how it is handled in the kidney and the drugs that cause hypomagnesaemia. The frequency and the number of drugs like diuretics and proton-pump inhibitors (PPIs) that are used daily in medical practice are discussed in order to prevent and treat adverse effects by providing an insight into Mg2+ homeostasis.
Collapse
Affiliation(s)
- Periklis Katopodis
- Department of Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, London, UB8 3PH, UK.
| | - Emmanouil Karteris
- Department of Biosciences, College of Health and Life Sciences, Brunel University London, Uxbridge, London, UB8 3PH, UK
| | | |
Collapse
|
398
|
Schoch S, Sen V, Brenner W, Hartwig A, Köberle B. In Vitro Nephrotoxicity Studies of Established and Experimental Platinum-Based Compounds. Biomedicines 2021; 9:biomedicines9081033. [PMID: 34440237 PMCID: PMC8394219 DOI: 10.3390/biomedicines9081033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 11/16/2022] Open
Abstract
Cisplatin is one of the most commonly used drugs for the treatment of various solid cancers. However, its efficacy is restricted by severe side effects, especially dose-limiting nephrotoxicity. New platinum-based compounds are designed to overcome this limitation. Previous investigations showed that the platinum(IV)–nitroxyl complex PN149 is highly cytotoxic in various tumor cell lines. In the present study, investigations with PN149 were extended to normal human kidney tubule epithelia. Coincident with higher intracellular platinum accumulation, the cytotoxicity of PN149 in the proximal tubule epithelial cell line ciPTEC was more pronounced compared to the established platinum chemotherapeutics cisplatin, carboplatin and oxaliplatin. Quantitative gene expression profiling revealed the induction of ROS-inducible and anti-oxidative genes, suggesting an oxidative stress response by PN149. However, in contrast to cisplatin, no pro-inflammatory response was observed. Genes coding for distinct DNA damage response factors and genes related to apoptosis were up-regulated, indicating the activation of the DNA damage response system and induction of the apoptotic cascade by PN149. Altogether, a comparable transcriptional response was observed for PN149 and the platinum chemotherapeutics. However, the lack of inflammatory activity, which is a possible cause contributing to toxicity in human renal proximal tubule epithelia, might indicate the reduced nephrotoxic potential of PN149.
Collapse
Affiliation(s)
- Sarah Schoch
- Department of Laboratory Medicine, Lund University, Scheelevägen 2, 223 81 Lund, Sweden;
| | - Vasily Sen
- Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, 142432 Moscow, Russia;
| | - Walburgis Brenner
- Clinic for Obstetrics and Women’s Health, Department of Urology and Pediatric Urology, University Medical Center Mainz, Langenbeckstreet 1, 55131 Mainz, Germany;
| | - Andrea Hartwig
- Karlsruhe Institute of Technology, Department of Food Chemistry and Toxicology, Adenauerring 20, 76131 Karlsruhe, Germany;
| | - Beate Köberle
- Karlsruhe Institute of Technology, Department of Food Chemistry and Toxicology, Adenauerring 20, 76131 Karlsruhe, Germany;
- Correspondence: ; Tel.: +49-721-608-42933
| |
Collapse
|
399
|
Brau-Figueroa H, Arango-Bravo E, Castro-Eguiluz D, Galicia-Carmona T, Lugo-Alferez LA, Cruz-Bautista I, Jiménez-Lima R, Cetina-Pérez L. Effectiveness of Concomitant Chemoradiotherapy with Gemcitabine in Locally Advanced Cervical Cancer Patients with Comorbidities. Cancer Res Treat 2021; 54:554-562. [PMID: 34384015 PMCID: PMC9016315 DOI: 10.4143/crt.2021.375] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 08/08/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose The standard treatment for locally advanced cervical cancer (LACC) is concomitant chemoradiotherapy with cisplatin (CDDP) followed by brachytherapy. The presence of comorbidities are risk factors for nephrotoxicity and are associated with lower survival. Gemcitabine is a radiosensitizing drug that has shown efficacy and safety in this context. The effectiveness of concomitant chemoradiotherapy with gemcitabine was evaluated versus cisplatin in LACC patients with comorbidities and preserved renal function. Materials and Methods An observational, longitudinal and paired study was carried out that included patients treated between February 2003 and December 2015. The primary objectives were to evaluate response rates, progression-free survival, and overall survival; the secondary objectives were to evaluate toxicity and renal function. Results Sixty-three patients treated with gemcitabine at 300 mg/m2 weekly and 126 patients treated with CDDP 40 mg/m2 weekly were included. There were no significant differences in response rates and survival rates. Treatment with cisplatin presented a higher frequency of hematological toxicities, while gemcitabine presented a higher frequency of gastrointestinal toxicities. A decrease in glomerular filtration rate (GFR; baseline vs. 1-year post-treatment) was observed in the cisplatin group (p=0.002), while not in the gemcitabine group (p=0.667). In a multivariate analysis, it is observed that only CDDP correlates with the decrease in GFR (hazard ratio, 2.42; p=0.012). Conclusion In LACC patients with comorbidities, gemcitabine and CDDP show the same efficacy, with different toxicity profiles. Treatment with cisplatin is associated with a significant decrease in GFR during follow-up, compared to treatment with gemcitabine that does not decrease it.
Collapse
Affiliation(s)
- Hasan Brau-Figueroa
- National Network of Cancer Records, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Eder Arango-Bravo
- Department of Clinical Research, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Denisse Castro-Eguiluz
- Consejo Nacional de Ciencia y Tecnología (CONACyT)- Department of Clinical Research, Instituto Nacional de Cancerología, Mexico City, Mexico
| | | | | | - Ivette Cruz-Bautista
- Research Unit in Metabolic Diseases, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Roberto Jiménez-Lima
- Department of Clinical Research, Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Lucely Cetina-Pérez
- Department of Clinical Research, Instituto Nacional de Cancerología, Mexico City, Mexico
| |
Collapse
|
400
|
Ghonaim E, El-Haggar S, Gohar S. Possible protective effect of pantoprazole against cisplatin-induced nephrotoxicity in head and neck cancer patients: a randomized controlled trial. Med Oncol 2021; 38:108. [PMID: 34357466 DOI: 10.1007/s12032-021-01558-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/27/2021] [Indexed: 12/17/2022]
Abstract
Cisplatin is used to treat solid malignancies including head and neck cancer. However, nephrotoxicity limits its use. In this study, we looked for a possible protective effect of pantoprazole against cisplatin-induced nephrotoxicity. We used novel biomarkers for early detection of nephrotoxicity. Sixty chemotherapy naïve head and neck cancer patients completed the study. Following complete history taking and thorough clinical examination, patients were randomly divided into three groups: 20 patients in each. Group I (control group) received cisplatin without pantoprazole, groups II and III received pantoprazole 80 mg and 40 mg, respectively, concurrently with cisplatin. Blood and urine samples were collected at baseline, and 48 h after the first and third cycles of cisplatin administration. Assessment of serum creatinine and soluble FasL (sFasL), as well as urinary neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1) was performed. Nephrotoxicity was detected in 6 patients in group I, none in group II and 3 patients in group III. Serum creatinine significantly increased at the end of treatment in group I compared to groups II and III. Group I also had significantly higher urinary KIM-1 and NGAL and serum sFasL compared to groups II and III after the first and third cycles. On the contrary, there was no significant difference between groups II and III. Pantoprazole prevented the increase in acute kidney injury biomarkers in cisplatin-treated patients. Therefore, it is a promising agent in reducing cisplatin-induced nephrotoxicity.Trial registration Clinical Trials.gov identifier: NCT04217512, registered in January 2020 " retrospectively registered".
Collapse
Affiliation(s)
- Eman Ghonaim
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, El-Guiesh Street, El-Gharbia Governorate, Tanta, 31111, Egypt.
| | - Sahar El-Haggar
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, El-Guiesh Street, El-Gharbia Governorate, Tanta, 31111, Egypt
| | - Suzy Gohar
- Oncology and Nuclear Medicine Department, Faculty of Medicine, Menoufia University, Yassin Abd-Elghaffar St. From Gamal Abdel Nasser ST., Shebin El-Kom, 32511, Menoufia, Egypt
| |
Collapse
|