1
|
Benjamin JI, Pati P, Luong T, Liu X, De Miguel C, Pollock JS, Pollock DM. Chronic mistimed feeding results in renal fibrosis and disrupted circadian blood pressure rhythms. Am J Physiol Renal Physiol 2024; 327:F683-F696. [PMID: 39205662 DOI: 10.1152/ajprenal.00047.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/12/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
Circadian disruption is a disturbance in biological timing, which can occur within or between different organizational levels, ranging from molecular rhythms within specific cells to the misalignment of behavioral and environmental cycles. Previous work from our group showed that less than 1 wk of food restriction to the light (inactive) period is sufficient to invert diurnal blood pressure rhythms in mice. However, kidney excretory rhythms and functions remained aligned with the light-dark cycle. Shift workers have an increased risk of cardiovascular disease that may different between sexes and often have irregular mealtimes, making the possibility of mistimed feeding as a potential contributor to the development of kidney disease. Thus, we hypothesized that chronic mistimed food intake would result in adverse cardiorenal effects, with sex differences in severity. Here, we show that chronic circadian disruption via mistimed feeding results in renal fibrosis and aortic stiffness in a sex-dependent manner. Our results indicate the importance of meal timing for the maintenance of blood pressure rhythms and kidney function, particularly in males. Our results also demonstrate that females are better able to acclimate to circadian-related behavioral change. NEW & NOTEWORTHY Circadian disruption through mistimed feeding resulted in nondipping blood pressure, renal fibrosis, and arterial stiffness that were less severe in females versus males. Mice fed exclusively during the daytime maintain their circadian rhythms of locomotor activity regardless of their loss of blood pressure rhythms. Although these mice ate less food, they maintained body weight, suggesting inefficiencies in overall metabolism. These findings demonstrate the importance of maintaining optimal food intake patterns to prevent cardiorenal pathophysiology.
Collapse
Affiliation(s)
- Jazmine I Benjamin
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Paramita Pati
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Tha Luong
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Xiaofen Liu
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Carmen De Miguel
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - Jennifer S Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| | - David M Pollock
- Section of Cardio-Renal Physiology and Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States
| |
Collapse
|
2
|
Zhang T, Widdop RE, Ricardo SD. Transition from acute kidney injury to chronic kidney disease: mechanisms, models, and biomarkers. Am J Physiol Renal Physiol 2024; 327:F788-F805. [PMID: 39298548 DOI: 10.1152/ajprenal.00184.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/15/2024] [Accepted: 09/01/2024] [Indexed: 09/22/2024] Open
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are increasingly recognized as interconnected conditions with overlapping pathophysiological mechanisms. This review examines the transition from AKI to CKD, focusing on the molecular mechanisms, animal models, and biomarkers essential for understanding and managing this progression. AKI often progresses to CKD due to maladaptive repair processes, persistent inflammation, and fibrosis, with both conditions sharing common pathways involving cell death, inflammation, and extracellular matrix (ECM) deposition. Current animal models, including ischemia-reperfusion injury (IRI) and nephrotoxic damage, help elucidate these mechanisms but have limitations in replicating the complexity of human disease. Emerging biomarkers such as kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and soluble tumor necrosis factor receptors (TNFRs) show promise in early detection and monitoring of disease progression. This review highlights the need for improved animal models and biomarker validation to better mimic human disease and enhance clinical translation. Advancing our understanding of the AKI-to-CKD transition through targeted therapies and refined research approaches holds the potential to significantly improve patient outcomes.
Collapse
Affiliation(s)
- Tingfang Zhang
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Robert E Widdop
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Sharon D Ricardo
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
3
|
Sone H, Lee TJ, Lee BR, Heo D, Oh S, Kwon SH. MicroRNA-mediated attenuation of branched-chain amino acid catabolism promotes ferroptosis in chronic kidney disease. Nat Commun 2023; 14:7814. [PMID: 38016961 PMCID: PMC10684653 DOI: 10.1038/s41467-023-43529-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 11/13/2023] [Indexed: 11/30/2023] Open
Abstract
Chronic kidney disease can develop from kidney injury incident to chemotherapy with cisplatin, which complicates the prognosis of cancer patients. MicroRNAs regulate gene expression by pairing with specific sets of messenger RNAs. Therefore, elucidating direct physical interactions between microRNAs and their target messenger RNAs can help decipher crucial biological processes associated with cisplatin-induced kidney injury. Through intermolecular ligation and transcriptome-wide sequencing, we here identify direct pairs of microRNAs and their target messenger RNAs in the kidney of male mice injured by cisplatin. We find that a group of cisplatin-induced microRNAs can target select messenger RNAs that affect the mitochondrial metabolic pathways in the injured kidney. Specifically, a cisplatin-induced microRNA, miR-429-3p, suppresses the pathway that catabolizes branched-chain amino acids in the proximal tubule, leading to cell death dependent on lipid peroxidation, called ferroptosis. Identification of miRNA-429-3p-mediated ferroptosis stimulation suggests therapeutic potential for modulating the branched-chain amino acid pathway in ameliorating cisplatin-induced kidney injury.
Collapse
Affiliation(s)
- Hisakatsu Sone
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Tae Jin Lee
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, 30912, USA
| | - Byung Rho Lee
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Dan Heo
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Sekyung Oh
- Department of Medical Science, Catholic Kwandong University College of Medicine, Incheon, 22711, South Korea
| | - Sang-Ho Kwon
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
4
|
Hucke A, Schröter R, Ceresa C, Chiorazzi A, Canta A, Semperboni S, Marmiroli P, Cavaletti G, Gess B, Ciarimboli G. Role of Mouse Organic Cation Transporter 2 for Nephro- and Peripheral Neurotoxicity Induced by Chemotherapeutic Treatment with Cisplatin. Int J Mol Sci 2023; 24:11486. [PMID: 37511245 PMCID: PMC10380567 DOI: 10.3390/ijms241411486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Cisplatin (CDDP) is an efficient chemotherapeutic agent broadly used to treat solid cancers. Chemotherapy with CDDP can cause significant unwanted side effects such as renal toxicity and peripheral neurotoxicity. CDDP is a substrate of organic cation transporters (OCT), transporters that are highly expressed in renal tissue. Therefore, CDDP uptake by OCT may play a role in causing unwanted toxicities of CDDP anticancer treatment. In this study, the contribution of the mouse OCT2 (mOCT2) to CDDP nephro- and peripheral neurotoxicity was investigated by comparing the effects of cyclic treatment with low doses of CDDP on renal and neurological functions in wild-type (WT) mice and mice with genetic deletion of OCT2 (OCT2-/- mice). This CDDP treatment protocol caused significant impairment of kidneys and peripherical neurological functions in WT mice. These effects were significantly reduced in OCT2-/- mice, however, less profoundly than what was previously measured in mice with genetic deletion of both OCT1 and 2 (OCT1-2-/- mice). Comparing the apparent affinities (IC50) of mOCT1 and mOCT2 for CDDP, the mOCT1 displayed a higher affinity for CDDP than the mOCT2 (IC50: 9 and 558 µM, respectively). Also, cellular toxicity induced by incubation with 100 µM CDDP was more pronounced in cells stably expressing mOCT1 than in cells expressing mOCT2. Therefore, in mice, CDDP uptake by both OCT1 and 2 contributes to the development of CDDP undesired side effects. OCT seem to be suitable targets for establishing treatment protocols aimed at decreasing unwanted CDDP toxicity and improving anticancer treatment with CDDP.
Collapse
Affiliation(s)
- Anna Hucke
- Experimentelle Nephrologie, Medizinische Klinik D, Universitätsklinikum Münster, 48149 Münster, Germany; (A.H.); (R.S.)
| | - Rita Schröter
- Experimentelle Nephrologie, Medizinische Klinik D, Universitätsklinikum Münster, 48149 Münster, Germany; (A.H.); (R.S.)
| | - Cecilia Ceresa
- Experimental Neurology Unit, Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy; (C.C.); (A.C.); (A.C.); (S.S.); (P.M.); (G.C.)
| | - Alessia Chiorazzi
- Experimental Neurology Unit, Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy; (C.C.); (A.C.); (A.C.); (S.S.); (P.M.); (G.C.)
| | - Annalisa Canta
- Experimental Neurology Unit, Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy; (C.C.); (A.C.); (A.C.); (S.S.); (P.M.); (G.C.)
| | - Sara Semperboni
- Experimental Neurology Unit, Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy; (C.C.); (A.C.); (A.C.); (S.S.); (P.M.); (G.C.)
| | - Paola Marmiroli
- Experimental Neurology Unit, Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy; (C.C.); (A.C.); (A.C.); (S.S.); (P.M.); (G.C.)
| | - Guido Cavaletti
- Experimental Neurology Unit, Department of Medicine and Surgery, University of Milano Bicocca, 20900 Monza, Italy; (C.C.); (A.C.); (A.C.); (S.S.); (P.M.); (G.C.)
- Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy
| | - Burkhard Gess
- Department of Neurology, University Hospital Münster, 48149 Münster, Germany;
- Department of Neurology, Evangelisches Klinikum Bethel, University of Bielefeld, 33617 Bielefeld, Germany
| | - Giuliano Ciarimboli
- Experimentelle Nephrologie, Medizinische Klinik D, Universitätsklinikum Münster, 48149 Münster, Germany; (A.H.); (R.S.)
| |
Collapse
|
5
|
Siddiqui NA, Ventrola AJ, Hartman AR, Konare T, Kamble NS, Thomas SC, Madaan T, Kharofa J, Sertorio MG, Kotagiri N. An Engineered Probiotic Platform for Cancer Epitope-Independent Targeted Radionuclide Therapy of Solid Tumors. Adv Healthc Mater 2023; 12:e2202870. [PMID: 36913614 PMCID: PMC10497710 DOI: 10.1002/adhm.202202870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/06/2023] [Indexed: 03/15/2023]
Abstract
Targeted radionuclide therapy (TRT) is an emerging therapeutic modality for the treatment of various solid cancers. Current approaches rely on the presence of cancer-specific epitopes and receptors against which a radiolabeled ligand is systemically administered to specifically deliver cytotoxic doses of α and β particles to tumors. In this proof-of-concept study, tumor-colonizing Escherichia coli Nissle 1917 (EcN) is utilized to deliver a bacteria-specific radiopharmaceutical to solid tumors in a cancer-epitope independent manner. In this microbe-based pretargeted approach, the siderophore-mediated metal uptake pathway is leveraged to selectively concentrate copper radioisotopes, 64 Cu and 67 Cu, complexed to yersiniabactin (YbT) in the genetically modified bacteria. 64 Cu-YbT facilitates positron emission tomography (PET) imaging of the intratumoral bacteria, whereas 67 Cu-YbT delivers a cytotoxic dose to the surrounding cancer cells. PET imaging with 64 Cu-YbT reveals persistence and sustained growth of the bioengineered microbes in the tumor microenvironment. Survival studies with 67 Cu-YbT reveals significant attenuation of tumor growth and extends survival of both MC38 and 4T1 tumor-bearing mice harboring the microbes. Tumor response to this pretargeted approach correlates with promising anti-tumor immunity, with noticeable CD8+ T:Treg cell ratio. Their strategy offers a pathway to target and ablate multiple solid tumors independent of their epitope and receptor phenotype.
Collapse
Affiliation(s)
- Nabil A. Siddiqui
- Division of Pharmaceutical SciencesJames L. Winkle College of PharmacyUniversity of CincinnatiCincinnatiOH45267USA
| | - Alec J. Ventrola
- Division of Pharmaceutical SciencesJames L. Winkle College of PharmacyUniversity of CincinnatiCincinnatiOH45267USA
| | - Alexandra R. Hartman
- Division of Pharmaceutical SciencesJames L. Winkle College of PharmacyUniversity of CincinnatiCincinnatiOH45267USA
| | - Tohonne Konare
- Division of Pharmaceutical SciencesJames L. Winkle College of PharmacyUniversity of CincinnatiCincinnatiOH45267USA
| | - Nitin S. Kamble
- Division of Pharmaceutical SciencesJames L. Winkle College of PharmacyUniversity of CincinnatiCincinnatiOH45267USA
| | - Shindu C. Thomas
- Division of Pharmaceutical SciencesJames L. Winkle College of PharmacyUniversity of CincinnatiCincinnatiOH45267USA
| | - Tushar Madaan
- Division of Pharmaceutical SciencesJames L. Winkle College of PharmacyUniversity of CincinnatiCincinnatiOH45267USA
| | - Jordan Kharofa
- Department of Radiation OncologyCollege of MedicineUniversity of CincinnatiCincinnatiOH45219USA
| | - Mathieu G. Sertorio
- Department of Radiation OncologyCollege of MedicineUniversity of CincinnatiCincinnatiOH45219USA
| | - Nalinikanth Kotagiri
- Division of Pharmaceutical SciencesJames L. Winkle College of PharmacyUniversity of CincinnatiCincinnatiOH45267USA
| |
Collapse
|
6
|
Ume AC, Wenegieme TY, Shelby JN, Paul-Onyia CDB, Waite AMJ, Kamau JK, Adams DN, Susuki K, Bennett ES, Ren H, Williams CR. Tacrolimus induces fibroblast-to-myofibroblast transition via a TGF-β-dependent mechanism to contribute to renal fibrosis. Am J Physiol Renal Physiol 2023; 324:F433-F445. [PMID: 36927118 PMCID: PMC10085566 DOI: 10.1152/ajprenal.00226.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 02/07/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023] Open
Abstract
Use of immunosuppressant calcineurin inhibitors (CNIs) is limited by irreversible kidney damage, hallmarked by renal fibrosis. CNIs directly damage many renal cell types. Given the diverse renal cell populations, additional targeted cell types and signaling mechanisms warrant further investigation. We hypothesized that fibroblasts contribute to CNI-induced renal fibrosis and propagate profibrotic effects via the transforming growth factor-β (TGF-β)/Smad signaling axis. To test this, kidney damage-resistant mice (C57BL/6) received tacrolimus (10 mg/kg) or vehicle for 21 days. Renal damage markers and signaling mediators were assessed. To investigate their role in renal damage, mouse renal fibroblasts were exposed to tacrolimus (1 nM) or vehicle for 24 h. Morphological and functional changes in addition to downstream signaling events were assessed. Tacrolimus-treated kidneys displayed evidence of renal fibrosis. Moreover, α-smooth muscle actin expression was significantly increased, suggesting the presence of fibroblast activation. TGF-β receptor activation and downstream Smad2/3 signaling were also upregulated. Consistent with in vivo findings, tacrolimus-treated renal fibroblasts displayed a phenotypic switch known as fibroblast-to-myofibroblast transition (FMT), as α-smooth muscle actin, actin stress fibers, cell motility, and collagen type IV expression were significantly increased. These findings were accompanied by concomitant induction of TGF-β signaling. Pharmacological inhibition of the downstream TGF-β effector Smad3 attenuated tacrolimus-induced phenotypic changes. Collectively, these findings suggest that 1) tacrolimus inhibits the calcineurin/nuclear factor of activated T cells axis while inducing TGF-β1 ligand secretion and receptor activation in renal fibroblasts; 2) aberrant TGF-β receptor activation stimulates Smad-mediated production of myofibroblast markers, notable features of FMT; and 3) FMT contributes to extracellular matrix expansion in tacrolimus-induced renal fibrosis. These results incorporate renal fibroblasts into the growing list of CNI-targeted cell types and identify renal FMT as a process mediated via a TGF-β-dependent mechanism.NEW & NOTEWORTHY Renal fibrosis, a detrimental feature of irreversible kidney damage, remains a sinister consequence of long-term calcineurin inhibitor (CNI) immunosuppressive therapy. Our study not only incorporates renal fibroblasts into the growing list of cell types negatively impacted by CNIs but also identifies renal fibroblast-to-myofibroblast transition as a process mediated via a TGF-β-dependent mechanism. This insight will direct future studies investigating the feasibility of inhibiting TGF-β signaling to maintain CNI-mediated immunosuppression while ultimately preserving kidney health.
Collapse
Affiliation(s)
- Adaku C Ume
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Tara Y Wenegieme
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Jennae N Shelby
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Chiagozie D B Paul-Onyia
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Aston M J Waite
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - John K Kamau
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Danielle N Adams
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Keiichiro Susuki
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Eric S Bennett
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Hongmei Ren
- Department of Biochemistry and Molecular Biology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| | - Clintoria R Williams
- Department of Neuroscience, Cell Biology, and Physiology, Boonshoft School of Medicine and College of Science and Mathematics, Wright State University, Dayton, Ohio, United States
| |
Collapse
|
7
|
Hodgson R, Crockford TL, Bhandari A, Kepple JD, Back J, Cawthorne E, Abeler-Dörner L, Laing AG, Clare S, Speak A, Adams DJ, Dougan G, Hayday AC, Deobagkar-Lele M, Cornall RJ, Bull KR. Prolidase Deficiency Causes Spontaneous T Cell Activation and Lupus-like Autoimmunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:547-557. [PMID: 36637239 PMCID: PMC9946897 DOI: 10.4049/jimmunol.2200212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 12/10/2022] [Indexed: 01/14/2023]
Abstract
Prolidase deficiency (PD) is a multisystem disorder caused by mutations in the PEPD gene, which encodes a ubiquitously expressed metallopeptidase essential for the hydrolysis of dipeptides containing C-terminal proline or hydroxyproline. PD typically presents in childhood with developmental delay, skin ulcers, recurrent infections, and, in some patients, autoimmune features that can mimic systemic lupus erythematosus. The basis for the autoimmune association is uncertain, but might be due to self-antigen exposure with tissue damage, or indirectly driven by chronic infection and microbial burden. In this study, we address the question of causation and show that Pepd-null mice have increased antinuclear autoantibodies and raised serum IgA, accompanied by kidney immune complex deposition, consistent with a systemic lupus erythematosus-like disease. These features are associated with an accumulation of CD4 and CD8 effector T cells in the spleen and liver. Pepd deficiency leads to spontaneous T cell activation and proliferation into the effector subset, which is cell intrinsic and independent of Ag receptor specificity or antigenic stimulation. However, an increase in KLRG1+ effector CD8 cells is not observed in mixed chimeras, in which the autoimmune phenotype is also absent. Our findings link autoimmune susceptibility in PD to spontaneous T cell dysfunction, likely to be acting in combination with immune activators that lie outside the hemopoietic system but result from the abnormal metabolism or loss of nonenzymatic prolidase function. This knowledge provides insight into the role of prolidase in the maintenance of self-tolerance and highlights the importance of treatment to control T cell activation.
Collapse
Affiliation(s)
- Rose Hodgson
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Tanya L. Crockford
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Aneesha Bhandari
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jessica D. Kepple
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Jennifer Back
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Eleanor Cawthorne
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Adam G. Laing
- Department of Immunobiology, King’s College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom; and
| | - Simon Clare
- Wellcome Sanger Institute, Hinxton, United Kingdom
| | | | | | | | - Adrian C. Hayday
- Department of Immunobiology, King’s College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom; and
| | - Mukta Deobagkar-Lele
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Richard J. Cornall
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Katherine R. Bull
- MRC Human Immunology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
8
|
Orwick A, Sears SM, Sharp CN, Doll MA, Shah PP, Beverly LJ, Siskind LJ. Lung cancer-kidney cross talk induces kidney injury, interstitial fibrosis, and enhances cisplatin-induced nephrotoxicity. Am J Physiol Renal Physiol 2023; 324:F287-F300. [PMID: 36727944 PMCID: PMC9988526 DOI: 10.1152/ajprenal.00317.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Patients with cancer represent a unique patient population with increased susceptibility to kidney disease. Drug-induced acute kidney injury (AKI) in patients with cancer is a common problem. Cisplatin is a highly effective treatment used in many solid-organ cancers and causes AKI in 30% of patients, increasing the risk of chronic kidney disease development. Most preclinical cisplatin toxicity studies have been completed in mice without cancer. We believe that the physiology of patients with cancer is not adequately represented in preclinical models, and the objective of this study was to determine how lung cancer will alter the nephrotoxicity of cisplatin. A genetically engineered mouse model and a syngeneic xenograft model of lung cancer were used. Mice were divided into the following four groups: 1) noncancer/vehicle, 2) noncancer/cisplatin, 3) cancer/vehicle, and 4) cancer/cisplatin. Mice were administered cisplatin via intraperitoneal injection once a week for 4 wk. Animals were euthanized 72 h following their final cisplatin injection. Mice with lung cancer had increased renal toxicity, injury, and fibrosis following repeated low doses of cisplatin. In addition, lung cancer alone induced kidney injury and fibrosis in the kidney before cisplatin treatment. In conclusion, this is the first study that we are aware of that assesses the impact of cancer on the kidney in conjunction with the nephrotoxicity of cisplatin. We believe that cancer is providing the first hit to the kidney and the subsequent damage from repeated doses of cisplatin becomes unsurmountable, leading to AKI and progression to chronic kidney disease.NEW & NOTEWORTHY Patients with cancer have impaired kidney function and increased susceptibility to nephrotoxic agents. Cisplatin is a commonly used chemotherapeutic with nephrotoxicity as the dose-limiting side effect. Cisplatin nephrotoxicity is almost exclusively studied in mice without cancer. Our current preclinical models do not adequately represent the complexity of patients with cancer. This study demonstrates increased renal toxicity, injury, and fibrosis in mice with lung cancer, which is exacerbated with cisplatin treatment. These results highlight the necessity of using preclinical models that more accurately capture the altered physiology of patients with cancer treated with cisplatin.
Collapse
Affiliation(s)
- Andrew Orwick
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
| | - Sophia M Sears
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
| | - Cierra N Sharp
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
| | - Mark A Doll
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
| | - Parag P Shah
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States
- Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States
| | - Levi J Beverly
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
- Department of Medicine, University of Louisville, Louisville, Kentucky, United States
- Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States
| | - Leah J Siskind
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky, United States
- Brown Cancer Center, University of Louisville, Louisville, Kentucky, United States
| |
Collapse
|
9
|
Shin SH, Wendland MF, Wang J, Velasquez M, Vandsburger MH. Noninvasively differentiating acute and chronic nephropathies via multiparametric urea-CEST, nuclear Overhauser enhancement-CEST, and quantitative magnetization transfer MRI. Magn Reson Med 2023; 89:774-786. [PMID: 36226662 PMCID: PMC11027791 DOI: 10.1002/mrm.29477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/26/2022] [Accepted: 09/14/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE Standardized blood tests often lack adequate sensitivity and specificity to capture the gradual progression of renal injuries. We suggest a multiparametric molecular MRI approach as a noninvasive tool for monitoring renal function loss and distinguishing different types of renal injuries. METHODS CEST and quantitative magnetization transfer (qMT) imaging were performed on cisplatin (n = 16) and aristolochic acid (AA)-induced nephropathy (n = 22) mouse models at 7T with an infusion of either saline or urea. Seven-pool Lorentzian fitting was applied for the analysis of CEST Z-spectra, and the T1 -corrected CEST contrast apparent exchange-dependent relaxation (AREX) from urea (+1 ppm) and two nuclear Overhauser enhancement (NOE) pools (-1.6 and -3.5 ppm) were measured. Similarly, qMT spectra were fitted into two-pool Ramani equation and the relative semi-solid macromolecular pool-size ratio was measured. Histology of mouse kidneys was performed to validate the MR findings. RESULTS AA model showed disrupted spatial gradients of urea in the kidney and significantly decreased NOE CEST and qMT contrast. The cisplatin model showed slightly decreased qMT contrast only. The orrelation of MR parameters to histological features showed that NOE CEST and qMT imaging are sensitive to both acute and chronic injuries, whereas urea CEST shows a significant correlation only to acute injuries. CONCLUSION These results indicate that our multiparametric approach allows comprehensive and totally noninvasive monitoring of renal function and histological changes for distinguishing different nephropathies.
Collapse
Affiliation(s)
- Soo Hyun Shin
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA
| | - Michael F. Wendland
- Berkeley Preclinical Imaging Core (BPIC), University of California, Berkeley, Berkeley, CA
| | - Jingshen Wang
- Department of Biostatistics, University of California, Berkeley, Berkeley, CA
| | - Mark Velasquez
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA
| | | |
Collapse
|
10
|
Cisplatin nephrotoxicity: new insights and therapeutic implications. Nat Rev Nephrol 2023; 19:53-72. [PMID: 36229672 DOI: 10.1038/s41581-022-00631-7] [Citation(s) in RCA: 139] [Impact Index Per Article: 139.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2022] [Indexed: 11/08/2022]
Abstract
Cisplatin is an effective chemotherapeutic agent for various solid tumours, but its use is limited by adverse effects in normal tissues. In particular, cisplatin is nephrotoxic and can cause acute kidney injury and chronic kidney disease. Preclinical studies have provided insights into the cellular and molecular mechanisms of cisplatin nephrotoxicity, which involve intracellular stresses including DNA damage, mitochondrial pathology, oxidative stress and endoplasmic reticulum stress. Stress responses, including autophagy, cell-cycle arrest, senescence, apoptosis, programmed necrosis and inflammation have key roles in the pathogenesis of cisplatin nephrotoxicity. In addition, emerging evidence suggests a contribution of epigenetic changes to cisplatin-induced acute kidney injury and chronic kidney disease. Further research is needed to determine how these pathways are integrated and to identify the cell type-specific roles of critical molecules involved in regulated necrosis, inflammation and epigenetic modifications in cisplatin nephrotoxicity. A number of potential therapeutic targets for cisplatin nephrotoxicity have been identified. However, the effects of renoprotective strategies on the efficacy of cisplatin chemotherapy needs to be thoroughly evaluated. Further research using tumour-bearing animals, multi-omics and genome-wide association studies will enable a comprehensive understanding of the complex cellular and molecular mechanisms of cisplatin nephrotoxicity and potentially lead to the identification of specific targets to protect the kidney without compromising the chemotherapeutic efficacy of cisplatin.
Collapse
|
11
|
Vichaya EG, Darpolor JK, Gross PS, Molkentine JM, Vermeer DW, Vermeer PD, Lee JH, Taniguchi CM, Dantzer R. Associative learning contributes to the persistence of fatigue-like behavior in male mice in a model of cancer survivorship. Brain Behav Immun 2023; 107:296-304. [PMID: 36323360 PMCID: PMC10208403 DOI: 10.1016/j.bbi.2022.10.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/15/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
Abstract
Persistent fatigue is a debilitating side effect that impacts a significant proportion of cancer survivors for which there is not yet an FDA-approved treatment. While certainly a multi-factorial problem, persistent fatigue could be due, in part, to associations learned during treatment. Therefore, we sought to investigate the role of associative learning in the persistence of fatigue using a preclinical model of cancer survivorship. For this purpose, we used a murine model of human papilloma virus-related head and neck cancer paired with a curative regimen of cisplatin-based chemoradiation in male C57BL/6J mice. Fatigue-like behavior was assessed by measuring variations in voluntary wheel running using a longitudinal design. Treatment robustly decreased voluntary wheel running, and this effect persisted for more than a month posttreatment. However, when wheels were removed during treatment, to minimize treatment-related fatigue, mice showed a more rapid return to baseline running levels. We confirmed that the delayed recovery observed in mice with continual wheel access was not due to increased treatment-related toxicity, in fact running attenuated cisplatin-induced kidney toxicity. Finally, we demonstrated that re-exposure to a treatment-related olfactory cue acutely re-instated fatigue. These data provide the first demonstration that associative processes can modulate the persistence of cancer-related fatigue-like behavior.
Collapse
Affiliation(s)
- Elisabeth G Vichaya
- Department of Psychology & Neuroscience, Baylor University, Waco, TX 76798, USA.
| | - Josephine K Darpolor
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center, Houston, TX 77030, USA
| | - Phillip S Gross
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jessica M Molkentine
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Daniel W Vermeer
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD 57104, USA
| | - Paola D Vermeer
- Cancer Biology Research Center, Sanford Research, Sioux Falls, SD 57104, USA
| | - John H Lee
- Avera Cancer Institute, 1000 E 23(rd) St., Sioux Falls, SD 57105, USA
| | - Cullen M Taniguchi
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
12
|
Ahmedy OA, El-Tanbouly DM, Al-Mokaddem AK, El-Said YA. Insights into the role of P2X7R/DUSP6/ERK1/2 and SIRT2/MDM2 signaling in the nephroprotective effect of berberine against cisplatin-induced renal fibrosis in rats. Life Sci 2022; 309:121040. [DOI: 10.1016/j.lfs.2022.121040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/26/2022] [Accepted: 10/01/2022] [Indexed: 11/16/2022]
|
13
|
Sears SM, Feng JL, Orwick A, Vega AA, Krueger AM, Shah PP, Doll MA, Beverly LJ, Siskind LJ. Pharmacological inhibitors of autophagy have opposite effects in acute and chronic cisplatin-induced kidney injury. Am J Physiol Renal Physiol 2022; 323:F288-F298. [PMID: 35796459 PMCID: PMC9394729 DOI: 10.1152/ajprenal.00097.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/31/2022] [Accepted: 06/08/2022] [Indexed: 12/25/2022] Open
Abstract
The nephrotoxicity of cisplatin remains a major hurdle in the field of oncology. Thirty percent of patients treated with cisplatin develop acute kidney injury, and all patients are at risk for long-term impacts on kidney function. There are currently no Federal Drug Administration-approved agents to prevent or treat cisplatin-induced kidney injury. The dosing regimen used in preclinical models of nephrotoxicity may impact the success of therapeutic candidates in clinical trials. Here, we demonstrated that pharmacological inhibitors of autophagy have opposite effects when used as interventions in two different models of cisplatin-induced kidney injury. Eight-week-old male C57BL/6 mice were treated with either one dose of 20 mg/kg cisplatin or weekly doses of 9 mg/kg cisplatin for 4 wk or until body weight loss exceeded 30%. Concurrently, mice were administered multiple doses of 60 mg/kg chloroquine or 15 mg/kg 3-methyladenine attempting to globally inhibit autophagy. Mice that received a single high dose of cisplatin had worsened kidney function, inflammation, and cell death with the addition of chloroquine. 3-Methlyadenine did not impact the development of acute kidney injury in this model. In contrast, mice that received repeated low doses of cisplatin showed improved kidney function, reduced inflammation, and reduced fibrosis when treated with either chloroquine or 3-methyladenine. This study highlights how therapeutic candidates can have drastically different effects on the development of cisplatin-induced kidney injury depending on the dosing model used. This emphasizes the importance of choosing the appropriate model of injury for preclinical studies.NEW & NOTEWORTHY This study examined how inhibition of autophagy has opposite effects on the development of acute and chronic kidney injury. Autophagy inhibition exacerbated the development of acute kidney injury following a single high dose of cisplatin but prevented the development of injury and fibrosis following repeated low doses of cisplatin.
Collapse
Affiliation(s)
- Sophia M Sears
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Joanna L Feng
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Andrew Orwick
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Alexis A Vega
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky
| | - Austin M Krueger
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Parag P Shah
- Department of Medicine, University of Louisville, Louisville, Kentucky
- Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Mark A Doll
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Levi J Beverly
- Department of Medicine, University of Louisville, Louisville, Kentucky
- Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Leah J Siskind
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
- Brown Cancer Center, University of Louisville, Louisville, Kentucky
| |
Collapse
|
14
|
Sears SM, Orwick A, Siskind LJ. Modeling Cisplatin-Induced Kidney Injury to Increase Translational Potential. Nephron Clin Pract 2022; 147:13-16. [PMID: 35793615 DOI: 10.1159/000525491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 06/01/2022] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Cisplatin-induced kidney injury is a major challenge hindering treatment of cancer patients. Thirty percent of patients treated with cisplatin develop acute kidney injury (AKI). Even patients that do not develop AKI are at risk for long-term decline in renal function and development of chronic kidney disease. Despite researchers' best efforts, no therapeutic agents to treat or prevent cisplatin-induced kidney injury have made it past phase 2 clinical trials. SUMMARY Modeling cisplatin-induced kidney injury in rodents has primarily been done using a single, high-dose model of injury. Newer models of injury have utilized repeated, low, or intermediate doses of cisplatin to incorporate the study of maladaptive repair processes following a renal insult. We believe that utilization of all these models is important to understand and treat the diverse types of cisplatin-induced kidney injury patients develop in the clinic. Incorporating comorbidities such as cancer and development of large animal models is also vital to increasing the human relevance of our studies. KEY MESSAGES Utilizing multiple dosing models of cisplatin-induced kidney injury, including relevant comorbidities and biological variables, and development of large animal models will increase the translational potential of preclinical studies.
Collapse
Affiliation(s)
- Sophia M Sears
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, Kentucky, USA
| | - Andrew Orwick
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, Kentucky, USA
| | - Leah J Siskind
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
15
|
Jia P, Xu S, Ren T, Pan T, Wang X, Zhang Y, Zou Z, Guo M, Zeng Q, Shen B, Ding X. LncRNA IRAR regulates chemokines production in tubular epithelial cells thus promoting kidney ischemia-reperfusion injury. Cell Death Dis 2022; 13:562. [PMID: 35732633 PMCID: PMC9217935 DOI: 10.1038/s41419-022-05018-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/07/2022] [Accepted: 06/10/2022] [Indexed: 02/07/2023]
Abstract
Increasing evidence demonstrates that long noncoding RNAs (lncRNAs) play an important role in several pathogenic processes of the kidney. However, functions of lncRNAs in ischemic acute kidney injury (AKI) remain undefined. In this study, global lncRNA profiling indicated that many lncRNA transcripts were deregulated in kidney after ischemia reperfusion (IR). Among them, we identified IRAR (ischemia-reperfusion injury associated RNA) as a potential lncRNA candidate, which was mostly expressed by the tubular epithelial cells (TECs) after IR, involved in the development of AKI. GapmeR-mediated silencing and viral-based overexpression of IRAR were carried out to assess its function and contribution to IR-induced AKI. The results revealed that in vivo silencing of IRAR significantly reduced IR-induced proinflammatory cells infiltration and AKI. IRAR overexpression induced chemokine CCL2, CXCL1 and CXCL2 expression both in mRNA and protein levels in TECs, while, silencing of IRAR resulted in downregulation of these chemokines. RNA immunoprecipitation and RNA pulldown assay validated the association between IRAR and CCL2, CXCL1/2. Further examination revealed that specific ablation of CCL2 in TECs reduced macrophages infiltration and proinflammatory cytokine production, attenuated renal dysfunction in IR mice. Inhibition of CXC chemokine receptor 2 (receptor of CXCL1/2) reduced neutrofils infiltration, but had no overt effect on kidney function. To explore the mechanism of IRAR upregulation in kidney during IR, we analyzed promoter region of IRAR and predicted a potential binding site for transcription factor C/EBP β on IRAR promoter. Silencing of C/EBP β reduced IRAR expression in TECs. A dual-luciferase reporter assay and chromatin immunoprecipitation (ChIP) confirmed that IRAR was a transcriptional target of the C/EBP β. Altogether, our findings identify IRAR as a new player in the development of ischemic AKI through regulating chemokine production and immune cells infiltration, suggesting that IRAR is a potential target for prevention and/or attenuation of AKI.
Collapse
Affiliation(s)
- Ping Jia
- grid.8547.e0000 0001 0125 2443Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China ,Shanghai Medical Center of Kidney, Shanghai, China ,Kidney and Dialysis Institute of Shanghai, Shanghai, China ,Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China
| | - Sujuan Xu
- grid.8547.e0000 0001 0125 2443Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ting Ren
- grid.8547.e0000 0001 0125 2443Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tianyi Pan
- grid.8547.e0000 0001 0125 2443Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaoyan Wang
- grid.8547.e0000 0001 0125 2443Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yunlu Zhang
- grid.8547.e0000 0001 0125 2443Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhouping Zou
- grid.8547.e0000 0001 0125 2443Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Man Guo
- grid.8547.e0000 0001 0125 2443Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qi Zeng
- grid.8547.e0000 0001 0125 2443Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bo Shen
- grid.8547.e0000 0001 0125 2443Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China ,Kidney and Dialysis Institute of Shanghai, Shanghai, China
| | - Xiaoqiang Ding
- grid.8547.e0000 0001 0125 2443Division of Nephrology, Zhongshan Hospital, Fudan University, Shanghai, China ,Shanghai Medical Center of Kidney, Shanghai, China ,Kidney and Dialysis Institute of Shanghai, Shanghai, China ,Kidney and Blood Purification Laboratory of Shanghai, Shanghai, China ,Hemodialysis quality control center of Shanghai, Shanghai, China
| |
Collapse
|
16
|
Sears SM, Vega AA, Kurlawala Z, Oropilla GB, Krueger A, Shah PP, Doll MA, Miller R, Beverly LJ, Siskind LJ. F4/80 hi Resident Macrophages Contribute to Cisplatin-Induced Renal Fibrosis. KIDNEY360 2022; 3:818-833. [PMID: 36128491 PMCID: PMC9438415 DOI: 10.34067/kid.0006442021] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 02/07/2022] [Indexed: 01/10/2023]
Abstract
Background Cisplatin-induced kidney injury remains a major obstacle in utilizing cisplatin as a chemotherapeutic for solid-organ cancers. Thirty percent of patients treated with cisplatin develop acute kidney injury (AKI), and even patients who do not develop AKI are at risk for long-term declines in kidney function and development of chronic kidney disease (CKD). Modeling cisplatin-induced kidney injury in mice has revealed that repeated low doses of cisplatin lead to development of kidney fibrosis. This model can be used to examine AKI-to-CKD transition processes. Macrophages play a role in some of these processes, including immune response, wound healing, and tissue remodeling. Depleting macrophage populations in the kidney reduced fibrosis development in other models of renal fibrosis. Methods We used either C57BL/6 mice with a Ccr2 genetic knockout or liposome encapsulated clodronate (Clodrosome) to deplete macrophage populations during repeated 9 mg/kg cisplatin treatments. We assessed how immune cell populations were altered in the blood and kidney of these mice and how these alterations affected development of renal fibrosis and kidney injury. Results We found that Clodrosome treatment decreased collagen deposition, myofibroblast accumulation, and inflammatory cytokine production, whereas Ccr2 genetic knockout had no effect on these markers after cisplatin treatment. Additionally, Ccr2-/- mice had decreased levels of F4/80lo infiltrating macrophages in the kidney after cisplatin treatments, but Clodrosome treatment depleted F4/80hi resident and CD206+ M2 macrophages. Conclusions These data suggest that Clodrosome depletion of F4/80hi and M2 macrophages in the kidney attenuates development of renal fibrosis after repeated low doses of cisplatin.
Collapse
Affiliation(s)
- Sophia M. Sears
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Alexis A. Vega
- Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky
| | - Zimple Kurlawala
- Department of Medicine, University of Louisville, Louisville, Kentucky
- Diabetes and Obesity Center, University of Louisville, Louisville, Kentucky
| | - Gabrielle B. Oropilla
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Austin Krueger
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Parag P. Shah
- Department of Medicine, University of Louisville, Louisville, Kentucky
- University of Louisville Brown Cancer Center, Louisville, Kentucky
| | - Mark A. Doll
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Robert Miller
- University of Louisville Brown Cancer Center, Louisville, Kentucky
- Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Levi J. Beverly
- Department of Medicine, University of Louisville, Louisville, Kentucky
- University of Louisville Brown Cancer Center, Louisville, Kentucky
| | - Leah J. Siskind
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
- University of Louisville Brown Cancer Center, Louisville, Kentucky
| |
Collapse
|
17
|
Bufi R, Korstanje R. The impact of genetic background on mouse models of kidney disease. Kidney Int 2022; 102:38-44. [DOI: 10.1016/j.kint.2022.03.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/07/2022] [Accepted: 03/14/2022] [Indexed: 12/24/2022]
|
18
|
Sears SM, Dupre TV, Shah PP, Davis DL, Doll MA, Sharp CN, Vega AA, Megyesi J, Beverly LJ, Snider AJ, Obeid LM, Hannun YA, Siskind LJ. Neutral ceramidase deficiency protects against cisplatin-induced acute kidney injury. J Lipid Res 2022; 63:100179. [PMID: 35151662 PMCID: PMC8953688 DOI: 10.1016/j.jlr.2022.100179] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/02/2022] [Accepted: 02/03/2022] [Indexed: 12/12/2022] Open
Abstract
Cisplatin is a commonly used chemotherapeutic for the treatment of many solid organ cancers; however, its effectiveness is limited by the development of acute kidney injury (AKI) in 30% of patients. AKI is driven by proximal tubule cell death, leading to rapid decline in renal function. It has previously been shown that sphingolipid metabolism plays a role in regulating many of the biological processes involved in cisplatin-induced AKI. For example, neutral ceramidase (nCDase) is an enzyme responsible for converting ceramide into sphingosine, which is then phosphorylated to become sphingosine-1-phosphate, and our lab previously demonstrated that nCDase knockout (nCDase-/-) in mouse embryonic fibroblasts led to resistance to nutrient and energy deprivation-induced cell death via upregulation of autophagic flux. In this study, we further characterized the role of nCDase in AKI by demonstrating that nCDase-/- mice are resistant to cisplatin-induced AKI. nCDase-/- mice display improved kidney function, reduced injury and structural damage, lower rates of apoptosis, and less ER stress compared to wild-type mice following cisplatin treatment. Although the mechanism of protection is still unknown, we propose that it could be mediated by increased autophagy, as chloroquine treatment resensitized nCDase-/- mice to AKI development. Taken together, we conclude that nCDase may represent a novel target to prevent cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Sophia M Sears
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA
| | - Tess V Dupre
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA
| | - Parag P Shah
- Department of Medicine, University of Louisville, Louisville, KY, USA; James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Deanna L Davis
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA
| | - Mark A Doll
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA
| | - Cierra N Sharp
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA
| | - Alexis A Vega
- Department of Biochemistry & Molecular Genetics, University of Louisville, Louisville, KY, USA
| | - Judit Megyesi
- Division of Nephrology, Department of Internal Medicine, University of Arkansas for Medical Sciences and Central Arkansas, Veterans Healthcare System, Little Rock, AR, USA
| | - Levi J Beverly
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA; Department of Medicine, University of Louisville, Louisville, KY, USA; James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
| | - Ashley J Snider
- Department of Nutritional Sciences, College of Agriculture and Life Sciences, University of Arizona, Tucson, AZ, USA
| | - Lina M Obeid
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA; Northport Veteran Affairs Medical Center, Northport, NY, USA
| | - Yusuf A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA; Northport Veteran Affairs Medical Center, Northport, NY, USA
| | - Leah J Siskind
- Department of Pharmacology & Toxicology, University of Louisville, Louisville, KY, USA; James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
19
|
Experimental models of acute kidney injury for translational research. Nat Rev Nephrol 2022; 18:277-293. [PMID: 35173348 DOI: 10.1038/s41581-022-00539-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 12/20/2022]
Abstract
Preclinical models of human disease provide powerful tools for therapeutic discovery but have limitations. This problem is especially apparent in the field of acute kidney injury (AKI), in which clinical trial failures have been attributed to inaccurate modelling performed largely in rodents. Multidisciplinary efforts such as the Kidney Precision Medicine Project are now starting to identify molecular subtypes of human AKI. In addition, over the past decade, there have been developments in human pluripotent stem cell-derived kidney organoids as well as zebrafish, rodent and large animal models of AKI. These organoid and AKI models are being deployed at different stages of preclinical therapeutic development. However, the traditionally siloed, preclinical investigator-driven approaches that have been used to evaluate AKI therapeutics to date rarely account for the limitations of the model systems used and have given rise to false expectations of clinical efficacy in patients with different AKI pathophysiologies. To address this problem, there is a need to develop more flexible and integrated approaches, involving teams of investigators with expertise in a range of different model systems, working closely with clinical investigators, to develop robust preclinical evidence to support more focused interventions in patients with AKI.
Collapse
|
20
|
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: 21] [Impact Index Per Article: 7.0] [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
|
21
|
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: 14] [Impact Index Per Article: 4.7] [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
|
22
|
Perše M. Cisplatin Mouse Models: Treatment, Toxicity and Translatability. Biomedicines 2021; 9:biomedicines9101406. [PMID: 34680523 PMCID: PMC8533586 DOI: 10.3390/biomedicines9101406] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/26/2021] [Accepted: 10/05/2021] [Indexed: 02/06/2023] Open
Abstract
Cisplatin is one of the most widely used chemotherapeutic drugs in the treatment of a wide range of pediatric and adult malignances. However, it has various side effects which limit its use. Cisplatin mouse models are widely used in studies investigating cisplatin therapeutic and toxic effects. However, despite numerous promising results, no significant improvement in treatment outcome has been achieved in humans. There are many drawbacks in the currently used cisplatin protocols in mice. In the paper, the most characterized cisplatin protocols are summarized together with weaknesses that need to be improved in future studies, including hydration and supportive care. As demonstrated, mice respond to cisplatin treatment in similar ways to humans. The paper thus aims to illustrate the complexity of cisplatin side effects (nephrotoxicity, gastrointestinal toxicity, neurotoxicity, ototoxicity and myelotoxicity) and the interconnectedness and interdependence of pathomechanisms among tissues and organs in a dose- and time-dependent manner. The paper offers knowledge that can help design future studies more efficiently and interpret study outcomes more critically. If we want to understand molecular mechanisms and find therapeutic agents that would have a potential benefit in clinics, we need to change our approach and start to treat animals as patients and not as tools.
Collapse
Affiliation(s)
- Martina Perše
- Medical Experimental Centre, Institute of Pathology, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
23
|
Wang S, Zhuang S, Dong Z. IFT88 deficiency in proximal tubular cells exaggerates cisplatin-induced injury by suppressing autophagy. Am J Physiol Renal Physiol 2021; 321:F269-F277. [PMID: 34251272 DOI: 10.1152/ajprenal.00672.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Primary cilia are widely regarded as specialized sensors in differentiated cells that have been implicated in the regulation of cell proliferation, differentiation, and viability. We have previously shown that shortening of primary cilia sensitizes cultured kidney tubular cells to cisplatin-induced apoptosis. Intraflagellar transport 88 (IFT88) is an essential component for ciliogenesis and maintenance. Here, we have further examined the effect of proximal tubule-specific IFT88 ablation on cisplatin-induced acute kidney injury (AKI). In this study, more severe AKI occurred in IFT88 knockout mice than age- and sex-matched wild-type mice. Mechanistically, cisplatin stimulated autophagy in kidney tubular cells as an intrinsic protective mechanism. However, renal autophagy was severely impaired in IFT88 knockout mice. In cultured HK-2 cells, cisplatin induced more apoptosis when IFT88 was knocked down. Tat-beclin 1 peptide, a specific autophagy activator, could partially prevent IFT88-associated cell death during cisplatin treatment, although cilium length was not improved significantly. Reexpression of IFT88 partially restored autophagy in IFT88 knockdown cells and suppressed apoptosis during cisplatin treatment. Taken together, these results indicate that defective autophagy in IFT88-deficient kidney cells and tissues contributes to the exaggerated AKI following cisplatin exposure.NEW & NOTEWORTHY Almost every cell has one hair-like, nonmotile antenna projecting from the cell surface, named the primary cilium. In kidney tubular cells, the primary cilium has a protective role, but the underlying mechanism is unclear. This study shows that a short cilium leads to the suppression of autophagy, which is responsible for the heightened injury sensitivity. These findings provide the clues of how to manipulate primary cilium and autophagy to save kidneys.
Collapse
Affiliation(s)
- Shixuan Wang
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| | - Shougang Zhuang
- Department of Medicine, Rhode Island Hospital and Alpert Medical School, Brown University, Providence, Rhode Island
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| |
Collapse
|
24
|
Sears SM, Siskind LJ. Potential Therapeutic Targets for Cisplatin-Induced Kidney Injury: Lessons from Other Models of AKI and Fibrosis. J Am Soc Nephrol 2021; 32:1559-1567. [PMID: 34049962 PMCID: PMC8425641 DOI: 10.1681/asn.2020101455] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/24/2021] [Accepted: 03/15/2021] [Indexed: 02/04/2023] Open
Abstract
The effectiveness of cisplatin, a mainstay in the treatment of many solid organ cancers, is hindered by dose-limiting nephrotoxicity. Cisplatin causes AKI in 30% of patients. Patients who do not develop AKI by clinical standards during treatment are still at risk for long-term decline in kidney function and the development of CKD. The connection between AKI and CKD has become increasingly studied, with renal fibrosis a hallmark of CKD development. To prevent both the short- and long-term effects of cisplatin, researchers must use models that reflect both types of pathology. Although a lot is known about cisplatin-induced AKI, very little is known about the mechanisms by which repeated low levels of cisplatin lead to fibrosis development. In this review, strategies used in various rodent models to prevent kidney injury, its progression to fibrosis, or both, are examined to gain mechanistic insights and identify potential therapeutic targets for cisplatin-induced kidney pathologies. Reviewing the results from these models highlights the diverse and highly complex role of cell death, cell senescence, endoplasmic reticulum stress, autophagy, and immune cell activation in acute and chronic kidney injuries. The use of several models of kidney injury is needed for development of agents that will prevent all aspects of cisplatin-induced kidney injury.
Collapse
Affiliation(s)
- Sophia M Sears
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
| | - Leah J Siskind
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, Kentucky
- James Graham Brown Cancer Center, Louisville, Kentucky
| |
Collapse
|
25
|
HIF in Nephrotoxicity during Cisplatin Chemotherapy: Regulation, Function and Therapeutic Potential. Cancers (Basel) 2021; 13:cancers13020180. [PMID: 33430279 PMCID: PMC7825709 DOI: 10.3390/cancers13020180] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/27/2020] [Accepted: 01/05/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Cisplatin is a widely used chemotherapy drug, but its use and efficacy are limited by its nephrotoxicity. HIF has protective effects against kidney injury during cisplatin chemotherapy, but it may attenuate the anti-cancer effect of cisplatin. In this review, we describe the role and regulation of HIF in cisplatin-induced nephrotoxicity and highlight the therapeutic potential of targeting HIF in chemotherapy. Abstract Cisplatin is a highly effective, broad-spectrum chemotherapeutic drug, yet its clinical use and efficacy are limited by its side effects. Particularly, cancer patients receiving cisplatin chemotherapy have high incidence of kidney problems. Hypoxia-inducible factor (HIF) is the “master” transcription factor that is induced under hypoxia to trans-activate various genes for adaptation to the low oxygen condition. Numerous studies have reported that HIF activation protects against AKI and promotes kidney recovery in experimental models of cisplatin-induced acute kidney injury (AKI). In contrast, little is known about the effects of HIF on chronic kidney problems following cisplatin chemotherapy. Prolyl hydroxylase (PHD) inhibitors are potent HIF inducers that recently entered clinical use. By inducing HIF, PHD inhibitors may protect kidneys during cisplatin chemotherapy. However, HIF activation by PHD inhibitors may reduce the anti-cancer effect of cisplatin in tumors. Future studies should test PHD inhibitors in tumor-bearing animal models to verify their effects in kidneys and tumors.
Collapse
|