1
|
Al-Marsoummi S, Singhal S, Garrett SH, Somji S, Sens DA, Singhal SK. CD133+CD24+ Renal Tubular Progenitor Cells Drive Hypoxic Injury Recovery via Hypoxia-Inducible Factor-1A and Epidermal Growth Factor Receptor Expression. Int J Mol Sci 2025; 26:2472. [PMID: 40141116 PMCID: PMC11942380 DOI: 10.3390/ijms26062472] [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: 01/19/2025] [Revised: 03/01/2025] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
CD133+CD24+ renal tubular progenitor cells play a crucial role in the repair and regeneration of renal tubules after acute kidney injury. The aim of this study is to investigate the responses of the human renal tubular precursor TERT (HRTPT) CD133+CD24+ cells and human renal epithelial cell 24 TERT (HREC24T) CD133-CD24+ cells to hypoxic stress, as well as their gene expression profiles. Whole transcriptome sequencing and functional network analysis identified distinct molecular characteristics of HRTPT cells as they were enriched with hypoxia-inducible factor-1A (HIF1A), epidermal growth factor (EGF), and endothelin-1 (EDN1). Our in vitro experiments demonstrated that, under hypoxia (2.5% oxygen), HRTPT cells showed minimal cell death and a 100-fold increase in HIF1A protein levels. In contrast, HREC24T cells exhibited significant cell death and only a two-fold increase in HIF1A protein level. These results indicate that CD133+CD24+ renal tubular progenitor cells have enhanced survival mechanisms under hypoxic stress, enabling them to survive and proliferate to replace damaged tubular cells. This study provides novel insights into the protective role of CD133+CD24+ renal tubular progenitor cells in hypoxic renal injury and identifies their potential survival mechanisms.
Collapse
Affiliation(s)
- Sarmad Al-Marsoummi
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.-M.); (S.S.)
| | - Sonalika Singhal
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.-M.); (S.S.)
| | - Scott H. Garrett
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.-M.); (S.S.)
| | - Seema Somji
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.-M.); (S.S.)
| | - Donald A. Sens
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.-M.); (S.S.)
| | - Sandeep K. Singhal
- Department of Pathology, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58202, USA; (S.A.-M.); (S.S.)
- Bioinformatics Division Adjunct Faculty, Pathology and Cell Biology, Columbia University Medical Centre, New York, NY 10032, USA
| |
Collapse
|
2
|
Babicz RSE, Baylor N, Terlouw A, Faber DA, Fukushima K, Biondi RM, Bouley R, Brown D. Ribosomal s6 kinase is a mediator of aquaporin-2 S256 phosphorylation and membrane accumulation after EGFR inhibition with erlotinib. Am J Physiol Renal Physiol 2025; 328:F344-F359. [PMID: 39823198 DOI: 10.1152/ajprenal.00353.2024] [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: 12/11/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025] Open
Abstract
Vasopressin (VP) activates protein kinase A (PKA), resulting in phosphorylation events and membrane accumulation of aquaporin-2 (AQP2). Epidermal growth factor receptor (EGFR) inhibition with erlotinib also induces AQP2 membrane trafficking with a phosphorylation pattern similar to VP, but without increasing PKA activity. Here, we identify the ribosomal s6 kinase (RSK) as a major mediator phosphorylating AQP2 in this novel, erlotinib-induced pathway. We found that RSK was expressed in collecting duct principal cells in rat kidneys. RSK inhibition with BI-D1870 blocked erlotinib-induced AQP2 serine 256 (S256) phosphorylation and membrane accumulation. CRISPR-generated RSK knockout (KO) cells failed to show increased S256 phosphorylation in response to erlotinib. Like PKA, RSK was able to phosphorylate AQP2 S256 in vitro. Inhibition of phosphoinositide-dependent kinase-1 (PDK1), a known activator of RSK, blocked erlotinib-induced AQP2 S256 phosphorylation and membrane accumulation. We conclude that RSK is a crucial terminal kinase phosphorylating AQP2 at S256 upon EGFR inhibition by erlotinib.NEW & NOTEWORTHY Epidermal growth factor receptor (EGFR) inhibition with erlotinib induces aquaporin-2 (AQP2) membrane accumulation with a phosphorylation pattern similar to vasopressin (VP). Here, we identify the ribosomal s6 kinase (RSK) as a major mediator phosphorylating AQP2 in this novel, erlotinib-induced pathway. In addition, we show that phosphoinositide-dependent kinase-1 (PDK1), a known activator of RSK, is implicated in this pathway: PDK1 inhibition blocks erlotinib-induced AQP2 S256 phosphorylation and membrane accumulation.
Collapse
Affiliation(s)
- Richard S E Babicz
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Noah Baylor
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Abby Terlouw
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Daphne A Faber
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Kazuhiko Fukushima
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Ricardo M Biondi
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck SocietyBuenos AiresArgentina
| | - Richard Bouley
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Dennis Brown
- Division of Nephrology, Program in Membrane Biology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
3
|
Rhim WK, Woo J, Kim JY, Lee EH, Cha SG, Kim DS, Baek SW, Park CG, Kim BS, Kwon TG, Han DK. Multiplexed PLGA scaffolds with nitric oxide-releasing zinc oxide and melatonin-modulated extracellular vesicles for severe chronic kidney disease. J Adv Res 2025; 69:75-89. [PMID: 38537702 PMCID: PMC11954823 DOI: 10.1016/j.jare.2024.03.018] [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: 11/13/2023] [Revised: 03/15/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION With prevalence of chronic kidney disease (CKD) in worldwide, the strategies to recover renal function via tissue regeneration could provide alternatives to kidney replacement therapies. However, due to relatively low reproducibility of renal basal cells and limited bioactivities of implanted biomaterials along with the high probability of substance-inducible inflammation and immunogenicity, kidney tissue regeneration could be challenging. OBJECTIVES To exclude various side effects from cell transplantations, in this study, we have induced extracellular vesicles (EVs) incorporated cell-free hybrid PMEZ scaffolds. METHODS Hybrid PMEZ scaffolds incorporating essential bioactive components, such as ricinoleic acid grafted Mg(OH)2 (M), extracellular matrix (E), and alpha lipoic acid-conjugated ZnO (Z) based on biodegradable porous PLGA (P) platform was successfully manufactured. Consecutively, for functional improvements, melatonin-modulated extracellular vesicles (mEVs), derived from the human umbilical cord MSCs in chemically defined media without serum impurities, were also loaded onto PMEZ scaffolds to construct the multiplexed PMEZ/mEV scaffold. RESULTS With functionalities of Mg(OH)2 and extracellular matrix-loaded PLGA scaffolds, the continuous nitric oxide-releasing property of modified ZnO and remarkably upregulated regenerative functionalities of mEVs showed significantly enhanced kidney regenerative activities. Based on these, the structural and functional restoration has been practically achieved in 5/6 nephrectomy mouse models that mimicked severe human CKD. CONCLUSION Our study has proved the combinatory bioactivities of the biodegradable PLGA-based multiplexed scaffold for kidney tissue regeneration in 5/6 nephrectomy mouse representing a severe CKD model. The optimal microenvironments for the morphogenetic formations of renal tissues and functional restorations have successfully achieved the combinatory bioactivities of remarkable components for PMEZ/mEV, which could be a promising therapeutic alternative for CKD treatment.
Collapse
Affiliation(s)
- Won-Kyu Rhim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Jiwon Woo
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Jun Yong Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea; Department of Biomedical Engineering and Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea; Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Eun Hye Lee
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea
| | - Seung-Gyu Cha
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea
| | - Seung-Woon Baek
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea; Department of Biomedical Engineering and Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea; Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering and Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea; Intelligent Precision of Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU) 2066 Seobu-ro, Jangan-gu, Suwon-si, Gyeonggi-do 16419, Republic of Korea
| | - Bum Soo Kim
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea; Department of Urology, School of Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea
| | - Tae Gyun Kwon
- Joint Institute for Regenerative Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea; Department of Urology, School of Medicine, Kyungpook National University, Jung-gu, Daegu 41944, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, 335 Pangyo-ro, Bundang-gu, Seongnam-si, Gyeonggi-do 13488, Republic of Korea.
| |
Collapse
|
4
|
Tito C, Masciarelli S, Colotti G, Fazi F. EGF receptor in organ development, tissue homeostasis and regeneration. J Biomed Sci 2025; 32:24. [PMID: 39966897 PMCID: PMC11837477 DOI: 10.1186/s12929-025-01119-9] [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: 06/21/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
The epidermal growth factor receptor (EGFR) is a protein embedded in the outer membrane of epithelial and mesenchymal cells, bone cells, blood and immune cells, heart cells, glia and stem neural cells. It belongs to the ErbB family, which includes three other related proteins: HER2/ErbB2/c-neu, HER3/ErbB3, and HER4/ErbB4. EGFR binds to seven known signaling molecules, including epidermal growth factor (EGF) and transforming growth factor-alpha (TGF-α). This binding triggers the formation of receptor pairs (dimers), self-phosphorylation of EGFR, and the activation of several signaling pathways within the cell. These pathways influence various cellular processes like proliferation, differentiation, migration, and survival. EGFR plays a critical role in both development and tissue homeostasis, including tissue repair and adult organ regeneration. Altered expression of EGFR is linked to disruption of tissue homeostasis and various diseases, among which cancer. This review focuses on how EGFR contributes to the development of different organs like the placenta, gut, liver, bone, skin, brain, T cell regulation, pancreas, kidneys, mammary glands and lungs along with their associated pathologies. The involvement of EGFR in organ-specific branching morphogenesis process is also discussed. The level of EGFR activity and its impact vary across different organs. Factors as the affinity of its ligands, recycling or degradation processes, and transactivation by other proteins or environmental factors (such as heat stress and smoking) play a role in regulating EGFR activity. Understanding EGFR's role and regulatory mechanisms holds promise for developing targeted therapeutic strategies.
Collapse
Affiliation(s)
- Claudia Tito
- Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology & Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14-16, 00161, Rome, Italy
| | - Silvia Masciarelli
- Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology & Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14-16, 00161, Rome, Italy
| | - Gianni Colotti
- Institute of Molecular Biology and Pathology, Italian National Research Council, IBPM-CNR, C/O Dept. Biochemical Sciences Sapienza University of Rome, Ed. CU027, P.Le A. Moro 5, 00185, Rome, Italy.
| | - Francesco Fazi
- Department of Anatomical, Histological, Forensic & Orthopaedic Sciences, Section of Histology & Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14-16, 00161, Rome, Italy.
| |
Collapse
|
5
|
Ezaddoustdar A, Kalina D, Bielohuby M, Boehm M, Wygrecka M. dEREGulated pathways: unraveling the role of epiregulin in skin, kidney, and lung fibrosis. Am J Physiol Cell Physiol 2025; 328:C617-C626. [PMID: 39750963 DOI: 10.1152/ajpcell.00813.2024] [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: 10/29/2024] [Revised: 12/24/2024] [Accepted: 12/24/2024] [Indexed: 01/04/2025]
Abstract
The epidermal growth factor receptor (EGFR) signaling pathway is an evolutionary conserved mechanism to control cell behavior during tissue development and homeostasis. Deregulation of this pathway has been associated with abnormal cell behavior, including hyperproliferation, senescence, and an inflammatory cell phenotype, thereby contributing to pathologies across a variety of organs, including the kidneys, skin, and lungs. To date, there are seven distinct EGFR ligands described. Although binding of these ligands to the receptor is cell type-specific and spatio-temporally controlled with distinct affinities and kinetics, epiregulin (EREG) stands out as a long-acting EGFR ligand that emerges under pathological conditions, particularly in tissue fibrosis. Although EREG has been extensively studied in cancer, its contribution to the maladaptive remodeling of tissue is elusive. The aim of this review is to highlight the role of EREG in skin, kidney, and lung fibrosis and to discuss opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Aysan Ezaddoustdar
- Center for Infection and Genomics of the Lung, Faculty of Medicine, Justus Liebig University, Member of the German Center for Lung Research, Giessen, Germany
| | | | | | | | - Malgorzata Wygrecka
- Center for Infection and Genomics of the Lung, Faculty of Medicine, Justus Liebig University, Member of the German Center for Lung Research, Giessen, Germany
- CSL Innovation GmbH, Marburg, Germany
- Institute for Lung Health, Justus Liebig University, Giessen, Germany
| |
Collapse
|
6
|
Tawengi M, Al-Dali Y, Tawengi A, Benter IF, Akhtar S. Targeting the epidermal growth factor receptor (EGFR/ErbB) for the potential treatment of renal pathologies. Front Pharmacol 2024; 15:1394997. [PMID: 39234105 PMCID: PMC11373609 DOI: 10.3389/fphar.2024.1394997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024] Open
Abstract
Epidermal growth factor receptor (EGFR), which is referred to as ErbB1/HER1, is the prototype of the EGFR family of receptor tyrosine kinases which also comprises ErbB2 (Neu, HER2), ErbB3 (HER3), and ErbB4 (HER4). EGFR, along with other ErbBs, is expressed in the kidney tubules and is physiologically involved in nephrogenesis and tissue repair, mainly following acute kidney injury. However, its sustained activation is linked to several kidney pathologies, including diabetic nephropathy, hypertensive nephropathy, glomerulonephritis, chronic kidney disease, and renal fibrosis. This review aims to provide a summary of the recent findings regarding the consequences of EGFR activation in several key renal pathologies. We also discuss the potential interplay between EGFR and the reno-protective angiotensin-(1-7) (Ang-(1-7), a heptapeptide member of the renin-angiotensin-aldosterone system that counter-regulates the actions of angiotensin II. Ang-(1-7)-mediated inhibition of EGFR transactivation might represent a potential mechanism of action for its renoprotection. Our review suggests that there is a significant body of evidence supporting the potential inhibition of EGFR/ErbB, and/or administration of Ang-(1-7), as potential novel therapeutic strategies in the treatment of renal pathologies. Thus, EGFR inhibitors such as Gefitinib and Erlinotib that have an acceptable safety profile and have been clinically used in cancer chemotherapy since their FDA approval in the early 2000s, might be considered for repurposing in the treatment of renal pathologies.
Collapse
Affiliation(s)
- Mohamed Tawengi
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Yazan Al-Dali
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| | | | - Ibrahim F Benter
- Faculty of Pharmacy, Final International University, Kyrenia, Cyprus
| | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
7
|
Mounieb F, Abdel-Sattar SA, Balah A, Akool ES. P2 X 7 receptor is a critical regulator of extracellular ATP-induced profibrotic genes expression in rat kidney: implication of transforming growth factor-β/Smad signaling pathway. Purinergic Signal 2024; 20:421-430. [PMID: 37934321 PMCID: PMC11303607 DOI: 10.1007/s11302-023-09977-4] [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: 07/06/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023] Open
Abstract
This study was designed to investigate the potential of extracellular adenosine 5'-triphosphate (ATP) via the P2 X 7 receptor to activate the renal fibrotic processes in rats. The present study demonstrates that administration of ATP rapidly activated transforming growth factor-β (TGF-β) to induce phosphorylation of Smad-2/3. Renal connective tissue growth factor (CTGF) and tissue inhibitor of metalloproteinase-1 (TIMP-1) mRNA and protein expressions were also increased following ATP administration. A decrease in TGF-β amount in serum as well as renal Smad-2/3 phosphorylation was noticed in animals pre-treated with the specific antagonist of P2 X 7 receptor, A 438,079. In addition, a significant reduction in mRNA and protein expression of CTGF and TIMP-1were also observed in the kidneys of those animals. Collectively, the current findings demonstrate that ATP has the ability to augment TGF-β-mediated Smad-2/3 phosphorylation and enhance the expression of the pro-fibrotic genes, CTGF and TIMP-1, an effect that is largely mediated via P2 X 7 receptor.
Collapse
Affiliation(s)
- Fatma Mounieb
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, 11751 El Nasr St, Nasr City, Cairo, Egypt
| | - Somaia A Abdel-Sattar
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, 11751 El Nasr St, Nasr City, Cairo, Egypt
| | - Amany Balah
- Pharmacology and Toxicology Department, Faculty of Pharmacy (Girls), Al-Azhar University, 11751 El Nasr St, Nasr City, Cairo, Egypt.
| | - El-Sayed Akool
- Pharmacology and Toxicology Department, Faculty of Pharmacy (boys), Al-Azhar University, Cairo, Egypt
| |
Collapse
|
8
|
Moreno J, Gluud LL, Galsgaard ED, Hvid H, Mazzoni G, Das V. Identification of ligand and receptor interactions in CKD and MASH through the integration of single cell and spatial transcriptomics. PLoS One 2024; 19:e0302853. [PMID: 38768139 PMCID: PMC11104622 DOI: 10.1371/journal.pone.0302853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/10/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Chronic Kidney Disease (CKD) and Metabolic dysfunction-associated steatohepatitis (MASH) are metabolic fibroinflammatory diseases. Combining single-cell (scRNAseq) and spatial transcriptomics (ST) could give unprecedented molecular disease understanding at single-cell resolution. A more comprehensive analysis of the cell-specific ligand-receptor (L-R) interactions could provide pivotal information about signaling pathways in CKD and MASH. To achieve this, we created an integrative analysis framework in CKD and MASH from two available human cohorts. RESULTS The analytical framework identified L-R pairs involved in cellular crosstalk in CKD and MASH. Interactions between cell types identified using scRNAseq data were validated by checking the spatial co-presence using the ST data and the co-expression of the communicating targets. Multiple L-R protein pairs identified are known key players in CKD and MASH, while others are novel potential targets previously observed only in animal models. CONCLUSION Our study highlights the importance of integrating different modalities of transcriptomic data for a better understanding of the molecular mechanisms. The combination of single-cell resolution from scRNAseq data, combined with tissue slide investigations and visualization of cell-cell interactions obtained through ST, paves the way for the identification of future potential therapeutic targets and developing effective therapies.
Collapse
Affiliation(s)
- Jaime Moreno
- Digital Science and Innovation, Computational Biology – AI & Digital Research, Novo Nordisk A/S, Maløv, Denmark
| | - Lise Lotte Gluud
- Gastro Unit, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
- Dept of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | | | - Henning Hvid
- Global Drug Discovery, Novo Nordisk A/S, Maløv, Denmark
| | - Gianluca Mazzoni
- Digital Science and Innovation, Computational Biology – AI & Digital Research, Novo Nordisk A/S, Maløv, Denmark
| | - Vivek Das
- Digital Science and Innovation, Computational Biology – AI & Digital Research, Novo Nordisk A/S, Maløv, Denmark
| |
Collapse
|
9
|
Forsse JS, Peterson MN, Papadakis Z, Taylor JK, Hess BW, Schwedock N, Allison DC, Griggs JO, Wilson RL, Grandjean PW. The Effect of Acute Aerobic Exercise on Biomarkers of Renal Health and Filtration in Moderate-CKD. RESEARCH QUARTERLY FOR EXERCISE AND SPORT 2024; 95:1-9. [PMID: 36608287 DOI: 10.1080/02701367.2022.2130131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 09/20/2022] [Indexed: 06/17/2023]
Abstract
Purpose: Efficacy of exercise to improve renal health and filtration remains understudied in adults with moderate-stages (stages G3a-b) of chronic kidney disease (CKD). Acute exercise may contribute clinically relevant information for exercise-related augmentation of renal health and filtration in CKD. Urine epidermal growth factor (uEGF) and cystatin C (CyC) are proposed to be more direct biomarkers of renal health and filtration. This study aimed to determine the influence of continuous moderate-intensity exercise (CMIE) and high-intensity interval exercise (HIIE) on traditional and novel biomarkers of renal health and filtration in moderate-stages of CKD. Methods: Twenty CKD participants completed 30 minutes of both CMIE and HIIE. Blood and urine samples were obtained pre, 1-hour, and 24-hours post-exercise. Traditional-serum creatinine (sCr) urine creatinine, novel-uEGF, uEGF ratio (uEGFr), and CyC. Estimates of glomerular filtration rate (eGFR)-modification of diet in renal disease (MDRD) and the CKD-Epidemiology (CKD-EPI)-responses were compared pre, 1 hr, and 24 hr post-exercise. Results: Relative to pre-exercise measures, uEGF remained unchanged in both exercise conditions. However, uEGFr was 5.4% greater 24-hours after HIIE (P = .05), while uEGFr remained unchanged with CMIE. sCr decreased 6 to 19% 1-hour post-exercise in both conditions (P = .009). On average renal filtration increased in eGFR-MDRD (7.2 ± 2.0 ml/min/1.73 m2) (P = .007) and eGFR-CKD-EPI (8.6 ± 2.3 ml/min/1.73 m2) 1-hour post-exercise (P = .009). Conclusion: By clinical estimates, renal filtration in CKD was not normalized but transiently improved regardless of exercise condition, with HIIE eliciting transient improvements in renal health.
Collapse
|
10
|
Kim M, Lee HH, Won SD, Jang Y, Kim BG, Cho NH, Choi YD, Chung JS, Han HH. Deciphering the Role of ERBB3 Isoforms in Renal Cell Carcinoma: A Comprehensive Genomic and Transcriptomic Analysis. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:181. [PMID: 38276060 PMCID: PMC10820170 DOI: 10.3390/medicina60010181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 01/17/2024] [Accepted: 01/19/2024] [Indexed: 01/27/2024]
Abstract
ERBB3, a key member of the receptor tyrosine kinase family, is implicated in the progression and development of various human cancers, affecting cellular proliferation and survival. This study investigated the expression of ERBB3 isoforms in renal clear cell carcinoma (RCC), utilizing data from 538 patients from The Cancer Genome Atlas (TCGA) Firehose Legacy dataset. Employing the SUPPA2 tool, the activity of 10 ERBB3 isoforms was examined, revealing distinct expression patterns in RCC. Isoforms uc001sjg.3 and uc001sjh.3 were found to have reduced activity in tumor tissues, while uc010sqb.2 and uc001sjl.3 demonstrated increased activity. These variations in isoform expression correlate with patient survival and tumor aggressiveness, indicating their complex role in RCC. The study, further, utilizes CIBERSORTx to analyze the association between ERBB3 isoforms and immune cell profiles in the tumor microenvironment. Concurrently, Gene Set Enrichment Analysis (GSEA) was applied, establishing a strong link between elevated levels of ERBB3 isoforms and critical oncogenic pathways, including DNA repair and androgen response. RT-PCR analysis targeting the exon 21-23 and exon 23 regions of ERBB3 confirmed its heightened expression in tumor tissues, underscoring the significance of alternative splicing and exon utilization in cancer development. These findings elucidate the diverse impacts of ERBB3 isoforms on RCC, suggesting their potential as diagnostic markers and therapeutic targets. This study emphasizes the need for further exploration into the specific roles of these isoforms, which could inform more personalized and effective treatment modalities for renal clear cell carcinoma.
Collapse
Affiliation(s)
- Mingyu Kim
- Center for Urologic Cancer, National Cancer Center, 323, Ilsan-Ro, Ilsandong-Gu, Goyang-Si 10408, Gyeonggi-Do, Republic of Korea; (M.K.); (H.H.L.); (S.D.W.)
| | - Hyung Ho Lee
- Center for Urologic Cancer, National Cancer Center, 323, Ilsan-Ro, Ilsandong-Gu, Goyang-Si 10408, Gyeonggi-Do, Republic of Korea; (M.K.); (H.H.L.); (S.D.W.)
| | - So Dam Won
- Center for Urologic Cancer, National Cancer Center, 323, Ilsan-Ro, Ilsandong-Gu, Goyang-Si 10408, Gyeonggi-Do, Republic of Korea; (M.K.); (H.H.L.); (S.D.W.)
| | - YeonSue Jang
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (Y.J.); (B.G.K.)
| | - Baek Gil Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (Y.J.); (B.G.K.)
| | - Nam Hoon Cho
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (Y.J.); (B.G.K.)
| | - Young Deuk Choi
- Department of Urology, Urological Science Institute, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea;
| | - Jin Soo Chung
- Center for Urologic Cancer, National Cancer Center, 323, Ilsan-Ro, Ilsandong-Gu, Goyang-Si 10408, Gyeonggi-Do, Republic of Korea; (M.K.); (H.H.L.); (S.D.W.)
| | - Hyun Ho Han
- Department of Urology, Urological Science Institute, Yonsei University College of Medicine, 50-1, Yonsei-Ro, Seodaemun-Gu, Seoul 03722, Republic of Korea;
| |
Collapse
|
11
|
Harskamp LR, Perez-Gomez MV, Heida JE, Engels GE, van Goor H, van den Heuvel MC, Streets AJ, Ong ACM, Ortiz A, Gansevoort RT. The association of urinary epidermal growth factors with ADPKD disease severity and progression. Nephrol Dial Transplant 2023; 38:2266-2275. [PMID: 36914219 PMCID: PMC10539218 DOI: 10.1093/ndt/gfad050] [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: 10/03/2022] [Indexed: 03/14/2023] Open
Abstract
BACKGROUND The epidermal growth factor receptor (EGFR) pathway is involved in kidney tissue repair and growth. Preclinical interventional data and scarce human data have suggested a role for this pathway in the pathophysiology of autosomal dominant polycystic kidney disease (ADPKD), while other data have suggested that its activation is causally linked to repair of damaged kidney tissue. We hypothesize that urinary EGFR ligands, as a reflection of EGFR activity, are associated with kidney function decline in ADPKD in the context of tissue repair following injury, and as the disease progresses as a sign of insufficient repair. METHODS In the present study, we measured the EGFR ligands, EGF and heparin binding-EGF (HB-EGF), in 24-h urine samples of 301 ADPKD patients and 72 age- and sex-matched living kidney donors to dissect the role of the EGFR pathway in ADPKD. During a median follow-up of 2.5 years, the association of urinary EGFR ligand excretion with annual change in estimated glomerular filtration rate (eGFR) and height-adjusted total kidney volume in ADPKD patients was analyzed using mixed-models methods, and the expression of three closely related EGFR family receptors in ADPKD kidney tissue was investigated by immunohistochemistry. Additionally, the effect of reducing renal mass (after kidney donation), was assessed to investigate whether urinary EGF matches this reduction and thus reflects the amount of remaining healthy kidney tissue. RESULTS At baseline, urinary HB-EGF did not differ between ADPKD patients and healthy controls (P = .6), whereas a lower urinary EGF excretion was observed in ADPKD patients [18.6 (11.8-27.8)] compared with healthy controls [51.0 (34.9-65.4) μg/24 h, P < .001]. Urinary EGF was positively associated with baseline eGFR (R = 0.54, P < .001) and a lower EGF was strongly associated with a more rapid GFR decline, even when adjusted for ADPKD severity markers (β = 1.96, P < .001), whereas HB-EGF was not. Expression of the EGFR, but not other EGFR-related receptors, was observed in renal cysts but was absent in non-ADPKD kidney tissue. Finally, unilateral nephrectomy resulted in a decrease of 46.4 (-63.3 to -17.6) % in urinary EGF excretion, alongside a decrease of 35.2 ± 7.2% in eGFR and 36.8 ± 6.9% in measured GFR (mGFR), whereas maximal mGFR (measured after dopamine induced hyperperfusion) decreased by 46.1 ± 7.8% (all P < .001). CONCLUSIONS Our data suggest that lower urinary EGF excretion may be a valuable novel predictor for kidney function decline in patients with ADPKD.
Collapse
Affiliation(s)
- Laura R Harskamp
- Department of Nephrology, University Medical Center of Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Judith E Heida
- Department of Nephrology, University Medical Center of Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Harry van Goor
- Department of Pathology and Medical Biology, University Medical Center of Groningen, University of Groningen, Groningen, The Netherlands
| | - Marius C van den Heuvel
- Department of Pathology and Medical Biology, University Medical Center of Groningen, University of Groningen, Groningen, The Netherlands
| | - Andrew J Streets
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Kidney Genetics Group, Academic Nephrology Unit, Sheffield, UK
| | - Albert C M Ong
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield Medical School, Kidney Genetics Group, Academic Nephrology Unit, Sheffield, UK
| | - Alberto Ortiz
- Department of Nephrology, Fundación Jiménez Díaz University Hospital and IIS-FJD, Madrid, Spain
| | - Ron T Gansevoort
- Department of Nephrology, University Medical Center of Groningen, University of Groningen, Groningen, The Netherlands
| |
Collapse
|
12
|
Kamianowska M, Rybi-Szumińska A, Kamianowska A, Maciejczyk M, Sołomianko K, Koput A, Wasilewska A. The Urinary Concentration of Trefoil Factor 3 (TFF3) in the Term and Preterm Neonates. J Clin Med 2023; 12:4936. [PMID: 37568337 PMCID: PMC10419516 DOI: 10.3390/jcm12154936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/13/2023] Open
Abstract
BACKGROUND Distinguishing between a pathologic state and renal development is important in neonatology. Because the assessment of serum creatinine in neonates is not reliable, better biomarkers are needed. Trefoil factor 3 (TFF3) is proposed as a biomarker of kidney injury. The study aimed to assess its urinary concentration in healthy term and stable preterm neonates. MATERIAL AND METHODS The study included 80 term and 20 preterm neonates born in the Department of Perinatology of the University Clinical Hospital in Bialystok. Urine was obtained from the term neonates on the 1st day of life and from the preterm neonates on the 1st, 8th, 15th and 22nd day of life. The urinary concentration of TFF3 was determined using a commercially available immunoassay and was normalized for the urinary creatinine concentration (cr.). RESULTS The values of TFF3/cr. were higher in the preterm than in the term neonates (p < 0.05) (median (Q1-Q3): 1486.85 (614.92-3559.18) and 317.29 (68.07-671.40) ng/mg cr.). They did not differ in the subsequent days of the preterm neonates' lives. The ROC curve for TFF3/cr. in the preterm and term neonates showed AUC = 0.751 (cut-off value = 1684.25 ng/mg cr.). CONCLUSIONS Prematurity is associated with higher urinary excretion of TFF3. Male gender is associated with an increased urinary TFF3 excretion in term neonates.
Collapse
Affiliation(s)
- Monika Kamianowska
- Department of Neonatology and Neonatal Intensive Care, Medical University of Bialystok, M. C. Sklodowskiej 24a Street, 15-276 Białystok, Poland;
| | - Agnieszka Rybi-Szumińska
- Department of Pediatrics and Nephrology, Medical University of Bialystok, 15-269 Bialystok, Poland (A.K.)
| | - Aleksandra Kamianowska
- Department of Pediatrics and Nephrology, Medical University of Bialystok, 15-269 Bialystok, Poland (A.K.)
| | - Mateusz Maciejczyk
- Department of Hygiene, Epidemiology and Ergonomic, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Katarzyna Sołomianko
- Department of Neonatology and Neonatal Intensive Care, Medical University of Bialystok, M. C. Sklodowskiej 24a Street, 15-276 Białystok, Poland;
| | - Alicja Koput
- Department of Pediatric Laboratory Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Anna Wasilewska
- Department of Pediatrics and Nephrology, Medical University of Bialystok, 15-269 Bialystok, Poland (A.K.)
| |
Collapse
|
13
|
Munguia-Galaviz FJ, Miranda-Diaz AG, Cardenas-Sosa MA, Echavarria R. Sigma-1 Receptor Signaling: In Search of New Therapeutic Alternatives for Cardiovascular and Renal Diseases. Int J Mol Sci 2023; 24:ijms24031997. [PMID: 36768323 PMCID: PMC9916216 DOI: 10.3390/ijms24031997] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Cardiovascular and renal diseases are among the leading causes of death worldwide, and regardless of current efforts, there is a demanding need for therapeutic alternatives to reduce their progression to advanced stages. The stress caused by diseases leads to the activation of protective mechanisms in the cell, including chaperone proteins. The Sigma-1 receptor (Sig-1R) is a ligand-operated chaperone protein that modulates signal transduction during cellular stress processes. Sig-1R interacts with various ligands and proteins to elicit distinct cellular responses, thus, making it a potential target for pharmacological modulation. Furthermore, Sig-1R ligands activate signaling pathways that promote cardioprotection, ameliorate ischemic injury, and drive myofibroblast activation and fibrosis. The role of Sig-1R in diseases has also made it a point of interest in developing clinical trials for pain, neurodegeneration, ischemic stroke, depression in patients with heart failure, and COVID-19. Sig-1R ligands in preclinical models have significantly beneficial effects associated with improved cardiac function, ventricular remodeling, hypertrophy reduction, and, in the kidney, reduced ischemic damage. These basic discoveries could inform clinical trials for heart failure (HF), myocardial hypertrophy, acute kidney injury (AKI), and chronic kidney disease (CKD). Here, we review Sig-1R signaling pathways and the evidence of Sig-1R modulation in preclinical cardiac and renal injury models to support the potential therapeutic use of Sig-1R agonists and antagonists in these diseases.
Collapse
Affiliation(s)
- Francisco Javier Munguia-Galaviz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Division de Ciencias de la Salud, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzman 49000, Jalisco, Mexico
| | - Alejandra Guillermina Miranda-Diaz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Miguel Alejandro Cardenas-Sosa
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Raquel Echavarria
- CONACYT-Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico
- Correspondence:
| |
Collapse
|
14
|
Cancer regulator EGFR-ErbB4 heterodimer is stabilized through glycans at the dimeric interface. J Mol Model 2022; 28:399. [DOI: 10.1007/s00894-022-05395-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022]
|
15
|
Li A, Ambruso SL, Oto OA, Barry M, Edelstein CL. A case report of pre-eclampsia-like endothelial injury in the kidney of an 85-year-old man treated with ibrutinib. BMC Nephrol 2022; 23:264. [PMID: 35870899 PMCID: PMC9308916 DOI: 10.1186/s12882-022-02873-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 07/03/2022] [Indexed: 01/29/2023] Open
Abstract
Abstract
Background
Glomerular endotheliosis is the pathognomonic glomerular lesion in pre-eclampsia that has also been described in those taking tyrosine kinase inhibitors for cancer treatment. Ibrutinib is a Bruton’s tyrosine kinase inhibitor used to treat chronic lymphocytic leukemia (CLL). We report the first known case of glomerular endotheliosis on kidney biopsy in a patient on ibrutinib monotherapy.
Case presentation
The patient presented with acute on chronic kidney disease, proteinuria, low C3 and C4 and a high rheumatoid factor titer. A kidney biopsy was performed to confirm a preliminary diagnosis of membranoproliferative glomerulonephritis (MPGN), the most common glomerular disease in patients with CLL. Unexpectedly, the kidney biopsy showed pre-eclampsia-like lesions on light and electron microscopy: occlusion of glomerular peripheral capillary lumens by swollen reactive endothelial cells. Findings of glomerulonephritis were not seen, and there were no specific glomerular immune deposits by immunofluorescence or electron microscopy.
Conclusions
CLL is known to cause glomerular lesions, mainly MPGN. There is increasing evidence that ibrutinib, a major treatment for CLL, can cause kidney disease, but the precise pathology is not characterized. We present a patient with CLL on ibrutinib with signs of glomerular endotheliosis. Based on the absence of CLL-induced kidney pathologies typically seen on the kidney biopsy and the non-selectivity of ibrutinib, we attributed the glomerular endotheliosis to ibrutinib. In pre-eclampsia, increased soluble fms-like tyrosine kinase 1 (sFlt1) levels induce endothelial dysfunction by decreasing vascular endothelial growth factor (VEGF). Ibrutinib has been demonstrated to have non-selective tyrosine kinase inhibition, including inhibition of VEGF receptor (VEGFR) and epidermal growth factor receptor (EGFR). VEGFR and EGFR inhibitors have recently been described in the literature to cause hypertension, proteinuria, and glomerular endotheliosis. Kidney biopsy should be performed in CLL patients on ibrutinib that present with acute kidney injury (AKI) or proteinuria to determine whether the clinical picture is attributable to the disease itself or a complication of the therapy.
Collapse
|
16
|
Urinary epidermal growth factor/monocyte chemotactic peptide 1 ratio as non-invasive predictor of Mayo clinic imaging classes in autosomal dominant polycystic kidney disease. J Nephrol 2022; 36:987-997. [DOI: 10.1007/s40620-022-01468-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 09/09/2022] [Indexed: 11/09/2022]
Abstract
Abstract
Background
Age- and height-adjusted total kidney volume is currently considered the best prognosticator in patients with autosomal dominant polycystic kidney disease. We tested the ratio of urinary epidermal growth factor and monocyte chemotactic peptide 1 for the prediction of the Mayo Clinic Imaging Classes.
Methods
Urinary epidermal growth factor and monocyte chemotactic peptide 1 levels were measured in two independent cohorts (discovery, n = 74 and validation set, n = 177) and healthy controls (n = 59) by immunological assay. Magnetic resonance imaging parameters were used for total kidney volume calculation and the Mayo Clinic Imaging Classification defined slow (1A–1B) and fast progressors (1C–1E). Microarray and quantitative gene expression analysis were used to test epidermal growth factor and monocyte chemotactic peptide 1 gene expression.
Results
Baseline ratio of urinary epidermal growth factor and monocyte chemotactic peptide 1 correlated with total kidney volume adjusted for height (r = − 0.6, p < 0.001), estimated glomerular filtration rate (r = 0.69 p < 0.001), discriminated between Mayo Clinic Imaging Classes (p < 0.001), and predicted the variation of estimated glomerular filtration rate at 10 years (r = − 0.51, p < 0.001). Conditional Inference Trees identified cut-off levels of the ratio of urinary epidermal growth factor and monocyte chemotactic peptide 1 for slow and fast progressors at > 132 (100% slow) and < 25.76 (89% and 86% fast, according to age), with 94% sensitivity and 66% specificity (p = 6.51E−16). Further, the ratio of urinary epidermal growth factor and monocyte chemotactic peptide 1 at baseline showed a positive correlation (p = 0.006, r = 0.36) with renal outcome (delta-estimated glomerular filtration rate per year, over a mean follow-up of 4.2 ± 1.2 years). Changes in the urinary epidermal growth factor and monocyte chemotactic peptide 1 were mirrored by gene expression levels in both human kidney cysts (epidermal growth factor: − 5.6-fold, fdr = 0.001; monocyte chemotactic peptide 1: 3.1-fold, fdr = 0.03) and Pkd1 knock-out mouse kidney (Egf: − 14.8-fold, fdr = 2.37E-20, Mcp1: 2.8-fold, fdr = 6.82E−15).
Conclusion
The ratio of urinary epidermal growth factor and monocyte chemotactic peptide 1 is a non-invasive pathophysiological biomarker that can be used for clinical risk stratification in autosomal dominant polycystic kidney disease.
Collapse
|
17
|
Harris RC. The Role of the Epidermal Growth Factor Receptor in Diabetic Kidney Disease. Cells 2022; 11:3416. [PMID: 36359813 PMCID: PMC9656309 DOI: 10.3390/cells11213416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 08/02/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) is expressed in numerous cell types in the adult mammalian kidney and is activated by a family of EGF-like ligands. EGFR activation has been implicated in a variety of physiologic and pathophysiologic functions. There is increasing evidence that aberrant EGFR activation is a mediator of progressive kidney injury in diabetic kidney disease. This review will highlight recent studies indicating its potential role and mechanisms of injury of both glomerular and tubular cells in development and progression of diabetic kidney disease.
Collapse
Affiliation(s)
- Raymond C. Harris
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt Center for Kidney Disease, Vanderbilt University Medical Center, Nashville, TN 37232, USA; ; Tel.: +1-615-202-9426
- Tennessee and Veterans Affairs, Nashville, TN 37232, USA
| |
Collapse
|
18
|
Massignam ET, Dieter C, Assmann TS, Duarte GCK, Bauer AC, Canani LH, Crispim D. The rs705708 A allele of the ERBB3 gene is associated with lower prevalence of diabetic retinopathy and arterial hypertension and with improved renal function in type 1 diabetic patients. Microvasc Res 2022; 143:104378. [PMID: 35594935 DOI: 10.1016/j.mvr.2022.104378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 04/21/2022] [Accepted: 05/11/2022] [Indexed: 11/29/2022]
Abstract
INTRODUCTION The Erb-b2 receptor tyrosine kinase 3 (ERBB3) is involved in autoimmune processes related to type 1 diabetes mellitus (T1DM) pathogenesis. Accordingly, some studies have suggested that single nucleotide polymorphisms (SNPs) in the ERBB3 gene confer risk for T1DM. Proliferation-associated protein 2G4 (PA2G4) is another candidate gene for this disease because it regulates cell proliferation and adaptive immunity. Moreover, PA2G4 regulates ERBB3. To date, no study has evaluated the association of PA2G4 SNPs and T1DM. AIM To evaluate the association of ERBB3 rs705708 (G/A) and PA2G4 rs773120 (C/T) SNPs with T1DM and its clinical and laboratory characteristics. METHODS This case-control study included 976 white subjects from Southern Brazil, categorized into 501 cases with T1DM and 475 non-diabetic controls. The ERBB3 and PA2G4 SNPs were genotyped by allelic discrimination-real-time PCR. RESULTS ERBB3 rs705708 and PA2G4 rs773120 SNPs were not associated with T1DM considering different inheritance models and also when controlling for covariables. However, T1DM patients carrying the ERBB3 rs705708 A allele developed T1DM at an earlier age vs. G/G patients. Interestingly, in the T1DM group, the rs705708 A allele was associated with lower prevalence of diabetic retinopathy and arterial hypertension as well as with improved renal function (higher estimated glomerular filtration rate and lower urinary albumin excretion levels) compared to G/G patients. CONCLUSIONS Although no association was observed between the ERBB3 rs705708 and PA2G4 rs773120 SNPs and T1DM, the rs705708 A allele was associated, for the first time in literature, with lower prevalence of diabetic retinopathy and arterial hypertension. Additionally, this SNP was associated with improved renal function.
Collapse
Affiliation(s)
- Eloísa Toscan Massignam
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, Rio Grande do Sul, Brazil
| | - Cristine Dieter
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, Rio Grande do Sul, Brazil
| | - Taís Silveira Assmann
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, Rio Grande do Sul, Brazil
| | - Guilherme Coutinho Kullmann Duarte
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, Rio Grande do Sul, Brazil
| | - Andrea Carla Bauer
- Nephrology Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Luis Henrique Canani
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, Rio Grande do Sul, Brazil
| | - Daisy Crispim
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil; Universidade Federal do Rio Grande do Sul, Faculty of Medicine, Department of Internal Medicine, Graduate Program in Medical Sciences: Endocrinology, Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
19
|
Forsse JS, Buckley D, Ismaeel A, Richardson KA, Oliver A, Koutakis P. Effect of Age and Acute-Moderate Intensity Exercise on Biomarkers of Renal Health and Filtration. BIOLOGY 2022; 11:527. [PMID: 35453726 PMCID: PMC9029611 DOI: 10.3390/biology11040527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 06/14/2023]
Abstract
Aerobic exercise elicits a multitude of physiological improvements in both healthy and diseased populations. However, acute changes in renal health and filtration with aerobic exercise remain difficult to quantify by traditional biomarkers to estimate glomerular filtration rate (eGFR). This study aimed to determine if an acute bout of moderate-intensity aerobic exercise transiently improves non-traditional biomarkers when compared to traditional biomarkers of renal health and filtration in individuals without cardiometabolic diseases. Thirty-nine participants (n = 18 men; n = 21 women; age 32.5 + 12.6 yr; height 171.1 + 11.4 cm; weight 78.7 + 15.6 kg; BMI 27.1 + 5.8) completed a single bout of moderate-intensity (50-60% HRR) aerobic exercise. Blood and urine samples were collected and compared before and post-exercise. Serum creatinine, urine epidermal growth factor (uEGF), uEGF/urine creatinine ratio (uEGFR), and cystatin C (CyC) were measured. In addition, eGFR-MDRD and the CKD-epidemiology equations were used to analyze renal clearance. Relative to pre-exercise measures: serum creatinine (p = 0.26), uEGF (p = 0.35), and uEGFR (p = 0.09) remained unchanged, whereas cystatin C (p = 0.00) significantly increased post-exercise. CyC eGFR was the only estimator of renal filtration to significantly change (p = 0.04). In conclusion, CyC is the only biomarker of renal health and filtration to significantly increase after aerobic exercise. Further investigation focused on sampling time and exercise-intensity is needed to solidify the current understanding of renal health and filtration.
Collapse
Affiliation(s)
- Jeffrey S. Forsse
- Department of Health Human Performance and Recreation, Baylor University, Waco, TX 76706, USA;
- Kinesiology Department, Stephen F. Austin State University, Nacogdoches, TX 75962, USA; (D.B.); (A.O.)
| | - David Buckley
- Kinesiology Department, Stephen F. Austin State University, Nacogdoches, TX 75962, USA; (D.B.); (A.O.)
- Integrative Immunology Laboratory, University of Texas Arlington, Arlington, TX 76019, USA
| | - Ahmed Ismaeel
- Clinical Muscle Biology Lab, Baylor University, Waco, TX 76706, USA;
| | - Kathleen A. Richardson
- Department of Health Human Performance and Recreation, Baylor University, Waco, TX 76706, USA;
| | - Autumn Oliver
- Kinesiology Department, Stephen F. Austin State University, Nacogdoches, TX 75962, USA; (D.B.); (A.O.)
- Edward Via College of Osteopathic Medicine, University of Louisiana Monroe, Monroe, LA 71203, USA
| | | |
Collapse
|
20
|
Tinel C, Lamarthée B, Callemeyn J, Van Loon E, Sauvaget V, Morin L, Aouni L, Rabant M, Gwinner W, Marquet P, Naesens M, Anglicheau D. Integrative Omics Analysis Unravels Microvascular Inflammation-Related Pathways in Kidney Allograft Biopsies. Front Immunol 2021; 12:738795. [PMID: 34795664 PMCID: PMC8593247 DOI: 10.3389/fimmu.2021.738795] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 10/15/2021] [Indexed: 12/29/2022] Open
Abstract
In solid-organ transplantation, microRNAs (miRNAs) have emerged as key players in the regulation of allograft cells function in response to injury. To gain insight into the role of miRNAs in antibody-mediated rejection, a rejection phenotype histologically defined by microvascular inflammation, kidney allograft biopsies were subjected to miRNA but also messenger RNA (mRNA) profiling. Using a unique multistep selection process specific to the BIOMARGIN study (discovery cohort, N=86; selection cohort, N=99; validation cohort, N=298), six differentially expressed miRNAs were consistently identified: miR-139-5p (down) and miR-142-3p/150-5p/155-5p/222-3p/223-3p (up). Their expression level gradually correlated with microvascular inflammation intensity. The cell specificity of miRNAs target genes was investigated by integrating their in vivo mRNA targets with single-cell RNA sequencing from an independent allograft biopsy cohort. Endothelial-derived miR-139-5p expression correlated negatively with MHC-related genes expression. Conversely, epithelial-derived miR-222-3p overexpression was strongly associated with degraded renal electrolyte homeostasis and repressed immune-related pathways. In immune cells, miR-150-5p regulated NF-κB activation in T lymphocytes whereas miR-155-5p regulated mRNA splicing in antigen-presenting cells. Altogether, integrated omics enabled us to unravel new pathways involved in microvascular inflammation and suggests that metabolism modifications in tubular epithelial cells occur as a consequence of antibody-mediated rejection, beyond the nearby endothelial compartment.
Collapse
Affiliation(s)
- Claire Tinel
- Necker-Enfants Malades Institute, Institut national de la santé et de la recherche médicale (Inserm) U1151, Université de Paris, Paris, France
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Baptiste Lamarthée
- Necker-Enfants Malades Institute, Institut national de la santé et de la recherche médicale (Inserm) U1151, Université de Paris, Paris, France
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Jasper Callemeyn
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Elisabet Van Loon
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Virginia Sauvaget
- Necker-Enfants Malades Institute, Institut national de la santé et de la recherche médicale (Inserm) U1151, Université de Paris, Paris, France
| | - Lise Morin
- Department of Nephrology and Kidney Transplantation, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Laïla Aouni
- Department of Nephrology and Kidney Transplantation, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Marion Rabant
- Necker-Enfants Malades Institute, Institut national de la santé et de la recherche médicale (Inserm) U1151, Université de Paris, Paris, France
- Department of Pathology, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Wilfried Gwinner
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| | - Pierre Marquet
- Institut national de la santé et de la recherche médicale (Inserm), University of Limoges, Limoges University Hospital, Pharmacology & Transplantation, Limoges, France
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Dany Anglicheau
- Necker-Enfants Malades Institute, Institut national de la santé et de la recherche médicale (Inserm) U1151, Université de Paris, Paris, France
- Department of Nephrology and Kidney Transplantation, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France
| |
Collapse
|
21
|
Lim CTS, M N, Sani D, Tan SN, Lim CW, Kirby BP, Ideris A, Stanslas J. Edible Bird Nest Protects the Kidney From Gentamicin Induced Acute Tubular Necrosis. Front Pharmacol 2021; 12:726005. [PMID: 34658864 PMCID: PMC8511955 DOI: 10.3389/fphar.2021.726005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/15/2021] [Indexed: 11/18/2022] Open
Abstract
Every year, there are about 13.3 million cases of acute kidney injury (AKI). Although AKI is a preventable and treatable disease, if left untreated, it has high risk of multiple organ failure and progression to end stage kidney disease. Acute tubular necrosis (ATN) has been recognised as one of the major causes of AKI. Till to date, there is no effective supplement or medication in treating or reversing AKI. Most of the treatment strategies involve preventative measure to minimise the occurrence of AKI or to reverse the cause of AKI. Hence one of the primary area of research interests is to explore the potential treatment for AKI. Edible bird nests (EBN) are edible food produce by the swiftlet’s saliva, which is rich in sialic acids. Sialic acids are monosaccharides that play a vital role in maintaining the integrity and proper function of the human organs, including kidneys. EBN also contains epidermal growth factor, which is widely believed to have rejuvenation and tissue repairing properties. We initiate this study to study the potential reno-protective effect of edible bird’s nests by studying the Wistar rat model of gentamicin-induced AKI. Besides renal profiles, renal histology was also semiquantitatively assessed. In our study, pre-treatment with EBN prevented and ameliorated the gentamicin-induced AKI. To a lesser extent, post-treatment with EBN also protected the kidney from the toxic effect of gentamicin. Our findings are highly indicative that EBN possesses reno-protective properties.
Collapse
Affiliation(s)
- Christopher T S Lim
- Department of Medicine, Faculty of Medicine and Health Sciences, UPM Serdang, Serdang, Malaysia
| | - Norhafizah M
- Department of Pathology, Faculty of Medicine and Health Sciences, UPM Serdang, Serdang, Malaysia
| | - D Sani
- Department of Medicine, Faculty of Medicine and Health Sciences, UPM Serdang, Serdang, Malaysia
| | - S N Tan
- Department of Medicine, Faculty of Medicine and Health Sciences, UPM Serdang, Serdang, Malaysia
| | - C W Lim
- Department of Medicine, Faculty of Medicine and Health Sciences, UPM Serdang, Serdang, Malaysia
| | - Brian P Kirby
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - A Ideris
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, UPM Serdang, Serdang, Malaysia
| | - J Stanslas
- Department of Medicine, Faculty of Medicine and Health Sciences, UPM Serdang, Serdang, Malaysia
| |
Collapse
|
22
|
Lin CT, Chen TH, Yang CC, Luo KH, Chen TH, Chuang HY. Epidermal Growth Factor Receptor (EGFR) Gene Polymorphism May be a Modifier for Cadmium Kidney Toxicity. Genes (Basel) 2021; 12:genes12101573. [PMID: 34680968 PMCID: PMC8535213 DOI: 10.3390/genes12101573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/25/2021] [Accepted: 10/01/2021] [Indexed: 11/16/2022] Open
Abstract
The results of many studies indicate that cadmium (Cd) exposure is harmful to humans, with the proximal tubule of the kidney being the main target of Cd accumulation and toxicity. Studies have also shown that Cd has the effect of activating the pathway of epidermal growth factor receptor (EGFR) signaling and cell growth. The EGFR is a family of transmembrane receptors, which are widely expressed in the human kidney. The aim of this study was to investigate the kidney function estimated glomerular filtration rate (eGFR), and its relationship with plasma Cd level and EGFR gene polymorphism. Using data from Academia Sinica Taiwan biobank, 489 subjects aged 30-70 years were analyzed. The demographic characteristics was determined from questionnaires, and biological sampling of urine and blood was determined from physical examination. Kidney function was assessed by the eGFR with CKD-EPI formula. Plasma Cd (ug/L) was measured by inductively coupled plasma mass spectrometry. A total of 97 single-nucleotide polymorphisms (SNPs) were identified in the EGFR on the Taiwan biobank chip, however 4 SNPs did not pass the quality control. Multiple regression analyses were performed to achieve the study aim. The mean (±SD) plasma Cd level of the study subjects was 0.02 (±0.008) ug/L. After adjusting for confounding variables, rs13244925 AA, rs6948867 AA, rs35891645 TT and rs6593214 AA types had higher eGFR (4.89 mL/min/1.73 m2 (p = 0.035), 5.54 mL/min/1.73 m2 (p = 0.03), 4.96 mL/min/1.73 m2 (p = 0.048) and 5.16 mL/min/1.73 m2 (p = 0.048), respectively). Plasma cadmium and rs845555 had an interactive effect on eGFR. In conclusion, EGFR polymorphisms could be modifiers of Cd kidney toxicity, in which rs13244925 AA, rs6948867 AA, rs35891645 TT and rs6593214 AA may be protective, and Cd interacting with rs845555 may affect kidney function.
Collapse
Affiliation(s)
- Chun-Ting Lin
- Department of Public, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-T.L.); (T.-H.C.)
| | - Ting-Hao Chen
- Department of Public, College of Health Sciences, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-T.L.); (T.-H.C.)
| | - Chen-Cheng Yang
- Departments of Occupational Medicine and Family Medicine, Kaohsiung Municipal Siaogang Hospital, and Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Kuei-Hau Luo
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Tzu-Hua Chen
- Department of Family Medicine, Kaohsiung Municipal Ta-Tung Hospital, and Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Hung-Yi Chuang
- Department of Occupational and Environmental Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Public Health and Environmental Medicine, College of Medicine, and Research Center for Environmental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-7312-1101 (ext. 6849)
| |
Collapse
|
23
|
Leierer J, Perco P, Hofer B, Eder S, Dzien A, Kerschbaum J, Rudnicki M, Mayer G. Coregulation Analysis of Mechanistic Biomarkers in Autosomal Dominant Polycystic Kidney Disease. Int J Mol Sci 2021; 22:6885. [PMID: 34206927 PMCID: PMC8269435 DOI: 10.3390/ijms22136885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disorder leading to deterioration of kidney function and end stage kidney disease (ESKD). A number of molecular processes are dysregulated in ADPKD but the exact mechanism of disease progression is not fully understood. We measured protein biomarkers being linked to ADPKD-associated molecular processes via ELISA in urine and serum in a cohort of ADPKD patients as well as age, gender and eGFR matched CKD patients and healthy controls. ANOVA and t-tests were used to determine differences between cohorts. Spearman correlation coefficient analysis was performed to assess coregulation patterns of individual biomarkers and renal function. Urinary epidermal growth factor (EGF) and serum apelin (APLN) levels were significantly downregulated in ADPKD patients. Serum vascular endothelial growth factor alpha (VEGFA) and urinary angiotensinogen (AGT) were significantly upregulated in ADPKD patients as compared with healthy controls. Arginine vasopressin (AVP) was significantly upregulated in ADPKD patients as compared with CKD patients. Serum VEGFA and VIM concentrations were positively correlated and urinary EGF levels were negatively correlated with urinary AGT levels. Urinary EGF and AGT levels were furthermore significantly associated with estimated glomerular filtration rate (eGFR) in ADPKD patients. In summary, altered protein concentrations in body fluids of ADPKD patients were found for the mechanistic markers EGF, APLN, VEGFA, AGT, AVP, and VIM. In particular, the connection between EGF and AGT during progression of ADPKD warrants further investigation.
Collapse
Affiliation(s)
- Johannes Leierer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| | - Paul Perco
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| | - Benedikt Hofer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| | - Susanne Eder
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| | | | - Julia Kerschbaum
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| | - Michael Rudnicki
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| | - Gert Mayer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria; (J.L.); (B.H.); (S.E.); (J.K.); (M.R.); (G.M.)
| |
Collapse
|
24
|
Shao G, Zhu S, Yang B. Applications of Herbal Medicine to Treat Autosomal Dominant Polycystic Kidney Disease. Front Pharmacol 2021; 12:629848. [PMID: 33986666 PMCID: PMC8111540 DOI: 10.3389/fphar.2021.629848] [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: 11/16/2020] [Accepted: 03/08/2021] [Indexed: 01/14/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a common hereditary kidney disease, which is featured by progressively enlarged bilateral fluid-filled cysts. Enlarging cysts destroy the structure of nephrons, ultimately resulting in the loss of renal function. Eventually, ADPKD develops into end-stage renal disease (ESRD). Currently, there is no effective drug therapy that can be safely used clinically. Patients progressed into ESRD usually require hemodialysis and kidney transplant, which is a heavy burden on both patients and society. Therefore, looking for effective therapeutic drugs is important for treating ADPKD. In previous studies, herbal medicines showed their great effects in multiple diseases, such as cancer, diabetes and mental disorders, which also might play a role in ADPKD treatment. Currently, several studies have reported that the compounds from herbal medicines, such as triptolide, curcumin, ginkolide B, steviol, G. lucidum triterpenoids, Celastrol, saikosaponin-d, Sparganum stoloniferum Buch.-Ham and Cordyceps sinensis, contribute to the inhibition of the development of renal cysts and the progression of ADPKD, which function by similar or different mechanisms. These studies suggest that herbal medicines could be a promising type of drugs and can provide new inspiration for clinical therapeutic strategy for ADPKD. This review summarizes the pharmacological effects of the herbal medicines on ADPKD progression and their underlying mechanisms in both in vivo and in vitro ADPKD models.
Collapse
Affiliation(s)
- Guangying Shao
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Shuai Zhu
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Baoxue Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China.,Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| |
Collapse
|
25
|
Comprehensive Biology and Genetics Compendium of Wilms Tumor Cell Lines with Different WT1 Mutations. Cancers (Basel) 2020; 13:cancers13010060. [PMID: 33379206 PMCID: PMC7801943 DOI: 10.3390/cancers13010060] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 12/01/2022] Open
Abstract
Simple Summary Wilms tumor is a childhood kidney tumor arising from embryonal cells. Wilms tumors are heterogeneous with several distinct subgroups that differ in their response to treatment. The genetic basis for these diverse forms of Wilms tumor is not fully understood. One subgroup of Wilms tumors is associated with mutations in the WT1 gene, encoding a transcription factor with a role in early kidney differentiation. Patients with WT1 mutant Wilms tumor may harbor germline mutations in this gene. Cell lines from Wilms tumors are notoriously difficult to establish and only few exist. We developed a method to cultivate cells from the WT1 mutant subtype of Wilms tumors and have established 11 cell lines with different mutations in WT1 to date. These cells will be instrumental to study the biology and genetics ultimately to develop precision treatments Abstract Purpose: WT1 mutant Wilms tumors represent a distinct subgroup, frequently associated with CTNNB1 mutations. The genetic basis for the development of this subtype is currently not fully understood. Methods: Live WT1 mutant Wilms tumors were collected during surgery of patients and cell cultures established in mesenchymal stem cell medium. They were studied for mutations in WT1 and CTNNB1, their differentiation capacity and protein activation status. Four cell lines were immortalized with a triple mutant ts SV40 largeT antigen and Telomerase. Results: 11 cell lines were established from Wilms tumors of nine patients, including a left and right tumor from the same patient and a primary and second tumor from another patient. Six patients had germ line and three were tumor specific mutations. All cell lines harbored only mutant or deleted WT1 genes. CTNNB1 was wild type in three, all others carried mutations affecting amino acid S45. They had variable and limited capacities for mesenchymal differentiation, a high migratory capacity and a low invasive potential. All cells showed an activation of multiple receptor tyrosine kinases and downstream signaling pathways. Conclusions: These cell lines represent an important new tool to study WT1 mutant Wilms tumors, potentially leading to new treatment approaches.
Collapse
|
26
|
de Castro-Suárez N, Trame MN, Ramos-Suzarte M, Dávalos JM, Bacallao-Mendez RA, Maceo-Sinabele AR, Mangas-Sanjuán V, Reynaldo-Fernández G, Rodríguez-Vera L. Semi-Mechanistic Pharmacokinetic Model to Guide the Dose Selection of Nimotuzumab in Patients with Autosomal Dominant Polycystic Kidney Disease. Pharmaceutics 2020; 12:pharmaceutics12121147. [PMID: 33256255 PMCID: PMC7760646 DOI: 10.3390/pharmaceutics12121147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 11/16/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a genetic disease characterized by an overexpression of epidermal growth factor receptor (EGFR). Nimotuzumab is a recombinant humanized monoclonal antibody against human EGFR. The aim of this study was to develop a population pharmacokinetic model for nimotuzumab and to identify demographic and clinical predictive factors of the pharmacokinetic variability. The population pharmacokinetics (PopPK) of nimotuzumab was characterized using a nonlinear mixed-effect modeling approach with NONMEM®. A total of 422 log-transformed concentration-versus-time datapoints from 20 patients enrolled in a single-center phase I clinical trial were used. Quasi steady state approximation of the full TMDD (target-mediated drug disposition) model with constant target concentration best described the concentration-time profiles. A turnover mediator was included which stimulates the non-specific clearance of mAb in the central compartment in order to explain the reduced levels at higher doses. Covariates had no influence on the PK (pharmacokinetics) parameters. The model was able to detect that the maximum effective dose in ADPKD subjects is 100 mg. The developed PopPK model may be used to guide the dose selection for nimotuzumab during routine clinical practice in patients with polycystic kidney disease. The model will further support the ongoing investigations of the PK/PD relationships of nimotuzumab to improve its therapeutic use in other disease areas.
Collapse
Affiliation(s)
- Niurys de Castro-Suárez
- Pharmacy Department, Institute of Food and Pharmacy, University of Havana, Havana 11300, Cuba; (N.d.C.-S.); (G.R.-F.); (L.R.-V.)
| | - Mirjam N. Trame
- AVROBIO Inc., Department of Translational Data Sciences and Advanced Analytics, Cambridge, MA 02139, USA;
| | | | - José M. Dávalos
- National Institute of Nephrology (INEF), Havana 10400, Cuba; (J.M.D.); (R.A.B.-M.)
| | | | | | - Víctor Mangas-Sanjuán
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, 46100 Valencia, Spain
- Interuniversity Research Institute for Molecular Recognition and Technological Development, Polytechnic University of Valencia, University of Valencia, 46100 Valencia, Spain
- Correspondence: ; Tel.: +3-49-6354-3351
| | - Gledys Reynaldo-Fernández
- Pharmacy Department, Institute of Food and Pharmacy, University of Havana, Havana 11300, Cuba; (N.d.C.-S.); (G.R.-F.); (L.R.-V.)
| | - Leyanis Rodríguez-Vera
- Pharmacy Department, Institute of Food and Pharmacy, University of Havana, Havana 11300, Cuba; (N.d.C.-S.); (G.R.-F.); (L.R.-V.)
| |
Collapse
|
27
|
Wang J, Tripathy N, Chung EJ. Targeting and therapeutic peptide-based strategies for polycystic kidney disease. Adv Drug Deliv Rev 2020; 161-162:176-189. [PMID: 32866560 PMCID: PMC7736157 DOI: 10.1016/j.addr.2020.08.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/15/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022]
Abstract
Polycystic kidney disease (PKD) is characterized by progressive cyst growth and is a leading cause of renal failure worldwide. Currently, there are limited therapeutic options available to PKD patients, and only one drug, tolvaptan, has been FDA-approved to slow cyst progression. Similar to other small molecule drugs, however, tolvaptan is costly, only moderately effective, and causes adverse events leading to high patient dropout rates. Peptides may mitigate many drawbacks of small molecule drugs, as they can be highly tissue-specific, biocompatible, and economically scaled-up. Peptides can function as targeting ligands that direct therapies to diseased renal tissue, or be potent as therapeutic agents themselves. This review discusses various aberrant signaling pathways in PKD and renal receptors that can be potential targets of peptide-mediated strategies. Additionally, peptides utilized in other kidney applications, but may prove useful in the context of PKD, are highlighted. Insights into novel peptide-based solutions that have potential to improve clinical management of PKD are provided.
Collapse
Affiliation(s)
- Jonathan Wang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Nirmalya Tripathy
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA; Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA; Department of Medicine, Division of Nephrology and Hypertension, University of Southern California, Los Angeles, CA, USA; Department of Surgery, Division of Vascular Surgery and Endovascular Therapy, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
28
|
Muñoz B, Schobel SA, Lisboa FA, Khatri V, Grey SF, Dente CJ, Kirk AD, Buchman T, Elster EA. Clinical risk factors and inflammatory biomarkers of post-traumatic acute kidney injury in combat patients. Surgery 2020; 168:662-670. [PMID: 32600883 DOI: 10.1016/j.surg.2020.04.064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 04/29/2020] [Accepted: 04/30/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Post-traumatic acute kidney injury has occurred in every major military conflict since its initial description during World War II. To ensure the proper treatment of combat casualties, early detection is critical. This study therefore aimed to investigate combat-related post-traumatic acute kidney injury in recent military conflicts, used machine learning algorithms to identify clinical and biomarker variables associated with the development of post-traumatic acute kidney injury, and evaluated the effects of post-traumatic acute kidney injury on wound healing and nosocomial infection. METHODS We conducted a retrospective clinical cohort review of 73 critically injured US military service members who sustained major combat-related extremity wounds and had collected injury characteristics, assayed serum and tissue biopsy samples for the expression of protein and messenger ribonucleic acid biomarkers. Bivariate analyses and random forest recursive feature elimination classification algorithms were used to identify associated injury characteristics and biomarker variables. RESULTS The incidence of post-traumatic acute kidney injury was 20.5%. Of that, 86% recovered baseline renal function and only 2 (15%) of the acute kidney injury group required renal replacement therapy. Random forest recursive feature elimination algorithms were able to estimate post-traumatic acute kidney injury with the area under the curve of 0.93, sensitivity of 0.91, and specificity of 0.91. Post-traumatic acute kidney injury was associated with injury severity score, serum epidermal growth factor, and tissue activin A type receptor 1, matrix metallopeptidase 10, and X-C motif chemokine ligand 1 expression. Patients with post-traumatic acute kidney injury exhibited poor wound healing and increased incidence of nosocomial infections. CONCLUSION The occurrence of acute kidney injury in combat casualties may be estimated using injury characteristics and serum and tissue biomarkers. External validations of these models are necessary to generalize for all trauma patients.
Collapse
Affiliation(s)
- Beau Muñoz
- Department of Surgery at the Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD; Uniformed Services University Surgical Critical Care Initiative, Bethesda, MD
| | - Seth A Schobel
- Department of Surgery at the Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD; Uniformed Services University Surgical Critical Care Initiative, Bethesda, MD; Henry Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD
| | - Felipe A Lisboa
- Department of Surgery at the Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD; Uniformed Services University Surgical Critical Care Initiative, Bethesda, MD; Henry Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD
| | - Vivek Khatri
- Department of Surgery at the Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD; Uniformed Services University Surgical Critical Care Initiative, Bethesda, MD; Henry Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD
| | - Scott F Grey
- Department of Surgery at the Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD; Uniformed Services University Surgical Critical Care Initiative, Bethesda, MD; Henry Jackson Foundation for the Advancement of Military Medicine, Inc, Bethesda, MD
| | - Christopher J Dente
- Uniformed Services University Surgical Critical Care Initiative, Bethesda, MD; Emory University, Atlanta, GA
| | - Allan D Kirk
- Uniformed Services University Surgical Critical Care Initiative, Bethesda, MD; Duke University, Durham, NC
| | - Timothy Buchman
- Uniformed Services University Surgical Critical Care Initiative, Bethesda, MD; Emory University, Atlanta, GA
| | - Eric A Elster
- Department of Surgery at the Uniformed Services University of the Health Sciences and the Walter Reed National Military Medical Center, Bethesda, MD; Uniformed Services University Surgical Critical Care Initiative, Bethesda, MD.
| |
Collapse
|
29
|
Kim BW, Kim SK, Heo KW, Bae KB, Jeong KH, Lee SH, Kim TH, Kim YH, Kang SW. Association between epidermal growth factor (EGF) and EGF receptor gene polymorphisms and end-stage renal disease and acute renal allograft rejection in a Korean population. Ren Fail 2020; 42:98-106. [PMID: 31906817 PMCID: PMC6968622 DOI: 10.1080/0886022x.2019.1710535] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Purpose Epidermal growth factor (EGF) has been found to be associated with the development and repair mechanisms of several renal diseases. In this study, we hypothesized that single nucleotide polymorphisms (SNPs) in EGF or its receptor genes might have an association with end-stage renal disease (ESRD) or acute renal allograft rejection (AR) in a Korean population. Methods Three-hundred and forty seven recipients of the first renal transplants for ESRD, including 63 AR patients along with 289 healthy adults were included in the study. Five EGF gene SNPs (rs11568835, rs11568943, rs2237051, rs11569017, and rs3756261) and four EGFR gene SNPs (rs1140475, rs2293347, rs1050171, and rs6965469) were analyzed. The genotypes of these SNPs were analyzed using the AxiomTM genome-wide human assay. Statistical analysis was performed using SNPStats and Haploview version 4.2 software. Multiple logistic regression models (codominant, dominant, recessive, and Log-additive) were used to estimate the odds ratio (OR), 95% confidence interval (CI), and P value. Results One SNP (rs11569017) in the EGF gene showed significant association with ESRD but not with AR. Another SNP (rs11568835) in the EGF gene showed significant association with susceptibility to AR but not with ESRD. One SNP (rs1050171) in the EGFR gene showed significant association with susceptibility to AR but not with ESRD. Conclusion Our findings suggest that SNPs in the EGF and EGFR gene may be associated with the risk of ESRD and AR development in the Korean population.
Collapse
Affiliation(s)
- Byeong Woo Kim
- Department of Internal Medicine, Haeundae Bumin Hospital, Busan, Korea
| | - Su Kang Kim
- Kohwang Medical Research Institute, Kyung Hee University School of Medicine, Seoul, Korea
| | - Kyung Wook Heo
- Department of Otolaryngology, Inje University, Busan, Korea
| | - Ki Beom Bae
- Department of General Surgery, Inje University, Busan, Korea
| | - Kyung Hwan Jeong
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, Korea
| | - Sang Ho Lee
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, Korea
| | - Tae Hee Kim
- Division of Nephrology, Department of Internal Medicine, Inje University, Busan, Korea
| | - Yeong Hoon Kim
- Division of Nephrology, Department of Internal Medicine, Inje University, Busan, Korea
| | - Sun Woo Kang
- Division of Nephrology, Department of Internal Medicine, Inje University, Busan, Korea
| |
Collapse
|
30
|
Azukaitis K, Schaefer F. Targeting Tubulointerstitium to Predict Kidney Outcomes in Childhood Nephrotic Syndrome. Kidney Int Rep 2020; 5:383-385. [PMID: 32281991 PMCID: PMC7136428 DOI: 10.1016/j.ekir.2020.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Karolis Azukaitis
- Clinic of Pediatrics, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Franz Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
31
|
Jian W, Wei CM, Guan JH, Mo CH, Xu YT, Zheng WB, Li L, Gui C. Association between serum HER2/ErbB2 levels and coronary artery disease: a case-control study. J Transl Med 2020; 18:124. [PMID: 32160892 PMCID: PMC7066824 DOI: 10.1186/s12967-020-02292-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 03/05/2020] [Indexed: 12/12/2022] Open
Abstract
Background Research has associated human epidermal growth factor receptor (HER2) with glucose and lipid metabolism. However, the association between circulating HER2 levels and coronary artery disease (CAD) remains to be elucidated. Methods We performed a case–control study with 435 participants (237 CAD patients and 198 controls) who underwent diagnostic coronary angiography from September 2018 to October 2019. Adjusted odds ratios (ORs) and 95% confidence intervals (CIs) for CAD were calculated with multiple logistic regression models after adjustment for confounders. Results Overall, increased serum HER2 levels were independently associated with the presence of CAD (OR per 1-standard deviation (SD) increase: 1.438, 95% CI 1.13–1.83; P = 0.003) and the number of stenotic vessels (OR per 1-SD increase: 1.399, 95% CI 1.15–1.71; P = 0.001). In the subgroup analysis, a significant interaction of HER2 with body mass index (BMI) on the presence of CAD was observed (adjusted interaction P = 0.046). Increased serum HER2 levels were strongly associated with the presence of CAD in participants with BMI ≥ 25 kg/m2 (OR per 1-SD increase: 2.143, 95% CI 1.37–3.35; P = 0.001), whereas no significant association was found in participants with BMI < 25 kg/m2 (OR per 1-SD increase: 1.225, 95% CI 0.90–1.67; P = 0.201). Conclusion Elevated HER2 level is associated with an increased risk of CAD, particularly in people with obesity. This finding yields new insight into the pathological mechanisms underlying CAD, and warrants further research regarding HER2 as a preventive and therapeutic target of CAD.
Collapse
Affiliation(s)
- Wen Jian
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, 06 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.,Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, 530021, Guangxi, People's Republic of China.,Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, 530021, Guangxi, People's Republic of China
| | - Chun-Mei Wei
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, 06 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.,Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, 530021, Guangxi, People's Republic of China.,Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, 530021, Guangxi, People's Republic of China
| | - Jia-Hui Guan
- Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, People's Republic of China
| | - Chang-Hua Mo
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, 06 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China
| | - Yu-Tao Xu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, 06 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.,Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, 530021, Guangxi, People's Republic of China.,Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, 530021, Guangxi, People's Republic of China
| | - Wen-Bo Zheng
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, 06 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.,Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, 530021, Guangxi, People's Republic of China.,Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, 530021, Guangxi, People's Republic of China
| | - Lang Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, 06 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China.,Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, 530021, Guangxi, People's Republic of China.,Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, 530021, Guangxi, People's Republic of China
| | - Chun Gui
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, 06 Shuangyong Road, Nanning, 530021, Guangxi, People's Republic of China. .,Guangxi Key Laboratory Base of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Nanning, 530021, Guangxi, People's Republic of China. .,Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, Nanning, 530021, Guangxi, People's Republic of China.
| |
Collapse
|
32
|
Parker MI, Nikonova AS, Sun D, Golemis EA. Proliferative signaling by ERBB proteins and RAF/MEK/ERK effectors in polycystic kidney disease. Cell Signal 2020; 67:109497. [PMID: 31830556 PMCID: PMC6957738 DOI: 10.1016/j.cellsig.2019.109497] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/05/2019] [Accepted: 12/06/2019] [Indexed: 12/24/2022]
Abstract
A primary pathological feature of polycystic kidney disease (PKD) is the hyperproliferation of epithelial cells in renal tubules, resulting in formation of fluid-filled cysts. The proliferative aspects of the two major forms of PKD-autosomal dominant PKD (ADPKD), which arises from mutations in the polycystins PKD1 and PKD2, and autosomal recessive PKD (ARPKD), which arises from mutations in PKHD1-has encouraged investigation into protein components of the core cell proliferative machinery as potential drivers of PKD pathogenesis. In this review, we examine the role of signaling by ERBB proteins and their effectors, with a primary focus on ADPKD. The ERBB family of receptor tyrosine kinases (EGFR/ERBB1, HER2/ERBB2, ERBB3, and ERBB4) are activated by extracellular ligands, inducing multiple pro-growth signaling cascades; among these, activation of signaling through the RAS GTPase, and the RAF, MEK1/2, and ERK1/2 kinases enhance cell proliferation and restrict apoptosis during renal tubuloepithelial cyst formation. Characteristics of PKD include overexpression and mislocalization of the ERBB receptors and ligands, leading to enhanced activation and increased activity of downstream signaling proteins. The altered regulation of ERBBs and their effectors in PKD is influenced by enhanced activity of SRC kinase, which is promoted by the loss of cytoplasmic Ca2+ and an increase in cAMP-dependent PKA kinase activity that stimulates CFTR, driving the secretory phenotype of ADPKD. We discuss the interplay between ERBB/SRC signaling, and polycystins and their depending signaling, with emphasis on thes changes that affect cell proliferation in cyst expansion, as well as the inflammation-associated fibrogenesis, which characterizes progressive disease. We summarize the current progress of preclinical and clinical trials directed at inhibiting this signaling axis, and discuss potential future strategies that may be productive for controlling PKD.
Collapse
Affiliation(s)
- Mitchell I Parker
- Program in Molecular Therapeutics, Fox Chase Cancer Center, 19111, USA; Molecular & Cell Biology & Genetics (MCBG) Program, Drexel University College of Medicine, 19102, USA
| | - Anna S Nikonova
- Program in Molecular Therapeutics, Fox Chase Cancer Center, 19111, USA
| | - Danlin Sun
- Program in Molecular Therapeutics, Fox Chase Cancer Center, 19111, USA; Institute of Life Science, Jiangsu University, Jingkou District, Zhenjiang, Jiangsu 212013, China
| | - Erica A Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, 19111, USA.
| |
Collapse
|
33
|
Zhu Q, Dong H, Bukhari AAS, Zhao A, Li M, Sun Y, Zhang X, Cao C, Su D, Liang X. HUWE1 promotes EGFR ubiquitination and degradation to protect against renal tubulointerstitial fibrosis. FASEB J 2020; 34:4591-4601. [PMID: 32017279 DOI: 10.1096/fj.201902751r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/08/2020] [Accepted: 01/18/2020] [Indexed: 12/13/2022]
Abstract
Injury of renal tubular epithelial cells is a key feature of the pathogenicity associated with tubulointerstitial fibrosis and other kidney diseases. HUWE1, an E3 ubiquitin ligase, acts by participating in ubiquitination and degradation of its target proteins. However, the detailed mechanisms by which HUWE1 might regulate fibrosis in renal tubular epithelial cells have not been established. Here, the possible regulation of renal tubulointerstitial fibrosis by HUWE1 was investigated by examining the expression of HUWE1 and EGFR in unilateral ureteral obstruction (UUO) mice. Markedly consistent reciprocal changes in HUWE1 and EGFR expression were observed at the protein and mRNA levels in the kidney after UUO injury. Expression of HUWE1 inhibited TGF-β-induced injury to HK-2 cells, while HUWE1 overexpression decreased the expression of EGFR. Further analysis indicated that HUWE1 physically interacted with EGFR and promoted its ubiquitination and degradation. HUWE1 expression also showed clinical relevance in renal disease, as it notably decreased in multiple types of clinical nephropathy, while EGFR expression significantly increased when compared to the normal kidney. Therefore, this study demonstrated that HUWE1, which serves as an E3 ubiquitin ligase specific for EGFR, promotes EGFR ubiquitination and degradation, thereby regulating EGFR expression and providing protection against kidney injury.
Collapse
Affiliation(s)
- Qian Zhu
- Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Hao Dong
- Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | | | - Anran Zhao
- Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Min Li
- Department of Pathology, Nanjing Medical University, Nanjing, China
| | - Yifei Sun
- Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Xue Zhang
- Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Changchun Cao
- Department of Nephrology, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, China
| | - Dongming Su
- Department of Pathology, Nanjing Medical University, Nanjing, China.,Center of Pathology and Clinical Laboratory, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, China
| | - Xiubin Liang
- Department of Pathophysiology, Nanjing Medical University, Nanjing, China.,Department of Nephrology, The Affiliated Sir Run Run Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
34
|
Al-Muhanna FA, Al-Rubaish AM, Vatte C, Mohiuddin SS, Cyrus C, Ahmad A, Shakil Akhtar M, Albezra MA, Alali RA, Almuhanna AF, Huang K, Wang L, Al-Kuwaiti F, Elsalamouni TSA, Al Hwiesh A, Huang X, Keating B, Li J, Lanktree MB, Al-Ali AK. Exome sequencing of Saudi Arabian patients with ADPKD. Ren Fail 2019; 41:842-849. [PMID: 31488014 PMCID: PMC6735335 DOI: 10.1080/0886022x.2019.1655453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Purpose: Autosomal dominant polycystic kidney disease (ADPKD) is characterized by progressive development of kidney cysts and enlargement and dysfunction of the kidneys. The Consortium of Radiologic Imaging Studies of the Polycystic Kidney Disease (CRISP) cohort revealed that 89.1% had either a PKD1 or PKD2 mutation. Of the CRISP patients with a genetic cause detected, mutations in PKD1 accounted for 85%, while mutations in the PKD2 accounted for the remaining 15%. Here, we report exome sequencing of 16 Saudi patients diagnosed with ADPKD and 16 ethnically matched controls. Methods: Exome sequencing was performed using combinatorial probe-anchor synthesis and improved DNA Nanoballs technology on BGISEQ-500 sequencers (BGI, China) using the BGI Exome V4 (59 Mb) Kit. Identified variants were validated with Sanger sequencing. Results: With the exception of GC-rich exon 1, we obtained excellent coverage of PKD1 (mean read depth = 88) including both duplicated and non-duplicated regions. Of nine patients with typical ADPKD presentations (bilateral symmetrical kidney involvement, positive family history, concordant imaging, and kidney function), four had protein truncating PKD1 mutations, one had a PKD1 missense mutation, and one had a PKD2 mutation. These variants have not been previously observed in the Saudi population. In seven clinically diagnosed ADPKD cases but with atypical features, no PKD1 or PKD2 mutations were identified, but rare predicted pathogenic heterozygous variants were found in cystogenic candidate genes including PKHD1, PKD1L3, EGF, CFTR, and TSC2. Conclusions: Mutations in PKD1 and PKD2 are the most common cause of ADPKD in Saudi patients with typical ADPKD. Abbreviations: ADPKD: Autosomal dominant polycystic kidney disease; CFTR: Cystic fibrosis transmembrane conductance regulator; EGF: Epidermal growth factor; MCIC: Mayo Clinic Imaging Classification; PKD: Polycystic kidney disease; TSC2: Tuberous sclerosis complex 2
Collapse
Affiliation(s)
- Fahad A Al-Muhanna
- Department of Internal Medicine, King Fahd Hospital of the University, Al-Khobar, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Abdullah M Al-Rubaish
- Department of Internal Medicine, King Fahd Hospital of the University, Al-Khobar, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Chittibabu Vatte
- Department of Clinical Biochemistry, College of Medicine, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Shamim Shaikh Mohiuddin
- Department of Clinical Biochemistry, College of Medicine, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Cyril Cyrus
- Department of Clinical Biochemistry, College of Medicine, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Arafat Ahmad
- Department of Clinical Biochemistry, College of Medicine, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Mohammed Shakil Akhtar
- Department of Clinical Biochemistry, College of Medicine, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | | | - Rudaynah A Alali
- Department of Internal Medicine, King Fahd Hospital of the University, Al-Khobar, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Afnan F Almuhanna
- Department of Radiology, King Fahd Hospital of the University, Al-Khobar, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Kai Huang
- BGI-Shenzhen , Shenzhen , China.,BGI-Shenzhen, China National GeneBank , Shenzhen , China
| | - Lusheng Wang
- Department of Computer Science, City University of Hong Kong , Hong Kong , Hong Kong
| | - Feras Al-Kuwaiti
- Department of Internal Medicine, King Fahd Hospital of the University, Al-Khobar, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Tamer S Ahmed Elsalamouni
- Department of Internal Medicine, King Fahd Hospital of the University, Al-Khobar, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Abdullah Al Hwiesh
- Department of Internal Medicine, King Fahd Hospital of the University, Al-Khobar, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| | - Xiaoyan Huang
- BGI-Shenzhen , Shenzhen , China.,BGI-Shenzhen, China National GeneBank , Shenzhen , China
| | - Brendan Keating
- Cardiovascular Institute, University of Pennsylvania School of Medicine , Philadelphia , PA , USA
| | - Jiankang Li
- BGI-Shenzhen , Shenzhen , China.,BGI-Shenzhen, China National GeneBank , Shenzhen , China.,Department of Computer Science, City University of Hong Kong , Hong Kong , Hong Kong
| | | | - Amein K Al-Ali
- Department of Clinical Biochemistry, College of Medicine, Imam Abdulrahman Bin Faisal University , Dammam , Saudi Arabia
| |
Collapse
|
35
|
Low levels of urinary epidermal growth factor predict chronic kidney disease progression in children. Kidney Int 2019; 96:214-221. [DOI: 10.1016/j.kint.2019.01.035] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 01/04/2019] [Accepted: 01/10/2019] [Indexed: 12/20/2022]
|
36
|
Yang L, Yuan H, Yu Y, Yu N, Ling L, Niu J, Gu Y. Epidermal growth factor receptor mimotope alleviates renal fibrosis in murine unilateral ureteral obstruction model. Clin Immunol 2019; 205:57-64. [PMID: 31152892 DOI: 10.1016/j.clim.2019.05.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/24/2019] [Accepted: 05/28/2019] [Indexed: 01/13/2023]
Abstract
Macrophages have been recognized as a vital factor that can promote renal fibrosis. Previously we reported that the EGFR mimotope could alleviate the macrophage infiltration in the Sjögren's syndrome-like animal model. In current study, we sought to observe whether the active immunization induced by the EGFR mimotope could ameliorate renal fibrosis in the murine Unilateral Ureteral Obstruction (UUO) model. A series of experiments showed the EGFR mimotope immunization could ameliorate renal fibrosis, reduce the expressions of fibronectin, α-SMA and collagen I and alleviate the infiltrations of F4/80+ macrophages in UUO model. Meanwhile, the EGFR mimotope immunization could inhibit the EGFR downstream signaling. Additionally, the frequency of and F4/80+CD9+/FAS+ macrophages significantly increased in spleen after the EGFR mimotope immunization. These evidence suggested that the EGFR mimotope could alleviate renal fibrosis by both inhibiting EGFR signaling and promoting macrophages apoptosis.
Collapse
Affiliation(s)
- Lin Yang
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Shanghai 200240, PR China
| | - Haoran Yuan
- Department of Central Laboratory, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Shanghai 200240, PR China
| | - Ying Yu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Shanghai 200240, PR China
| | - Nan Yu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Shanghai 200240, PR China
| | - Lilu Ling
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Shanghai 200240, PR China
| | - Jianying Niu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Shanghai 200240, PR China.
| | - Yong Gu
- Department of Nephrology, Shanghai Fifth People's Hospital, Fudan University, No. 801, Heqing Road, Shanghai 200240, PR China
| |
Collapse
|
37
|
Muiru AN, Shlipak MG, Scherzer R, Zhang WR, Ascher SB, Jotwani V, Grunfeld C, Parikh CR, Ng D, Palella FJ, Ho K, Kassaye S, Sharma A, Cohen M, Wang R, Qi Q, Estrella MM. Kidney disease risk factors associate with urine biomarkers concentrations in HIV-positive persons; a cross-sectional study. BMC Nephrol 2019; 20:4. [PMID: 30606136 PMCID: PMC6318986 DOI: 10.1186/s12882-018-1192-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/18/2018] [Indexed: 12/17/2022] Open
Abstract
Background HIV-positive persons bear an excess burden of chronic kidney disease (CKD); however, conventional methods to assess kidney health are insensitive and non-specific for detecting early kidney injury. Urinary biomarkers can detect early kidney injury, and may help mitigate the risk of overt CKD. Methods Cross-sectional study of HIV-positive persons in the Multicenter AIDS Cohort Study and the Women’s Interagency HIV Study. We measured levels of 14 biomarkers, capturing multiple dimensions of kidney injury. We then evaluated associations of known CKD risk factors with urine biomarkers using separate multivariable adjusted models for each biomarker. Results Of the 198 participants, one third were on HAART and virally suppressed. The vast majority (95%) had preserved kidney function as assessed by serum creatinine, with a median eGFR of 103 ml/min/1.73 m2 (interquartile range (IQR): 88, 116). In our multivariable analyses, the associations of each CKD risk factor with urinary biomarker levels varied in magnitude. For example, HIV viral load was predominantly associated with elevations in interleukin(IL)-18, and albuminuria, while higher CD4 levels were associated with lower monocyte chemoattractant protein-1 (MCP-1) and β2-microglobulin. In contrast, older age was significantly associated with elevations in α1-microglobulin, kidney injury marker-1, clusterin, MCP-1, and chitinase-3-like protein-1 levels, as well as lower epidermal growth factor, and uromodulin levels. Conclusions Among HIV-positive persons, CKD risk factors are associated with unique and heterogeneous patterns of changes in urine biomarkers levels. Additional work is needed to develop parsimonious algorithms that integrate multiple biomarkers and clinical data to discern the risk of overt CKD and its progression. Electronic supplementary material The online version of this article (10.1186/s12882-018-1192-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anthony N Muiru
- Kidney Health Research Collaborative, Department of Medicine, San Francisco Veterans Affairs Medical Center and University of California, 533 Parnassus Avenue, U404, Box 0532, San Francisco, CA, 94143, USA.
| | - Michael G Shlipak
- Kidney Health Research Collaborative, Department of Medicine, San Francisco Veterans Affairs Medical Center and University of California, 533 Parnassus Avenue, U404, Box 0532, San Francisco, CA, 94143, USA.,Department Epidemiology, and Biostatistics, University of California, San Francisco, CA, USA
| | - Rebecca Scherzer
- Kidney Health Research Collaborative, Department of Medicine, San Francisco Veterans Affairs Medical Center and University of California, 533 Parnassus Avenue, U404, Box 0532, San Francisco, CA, 94143, USA
| | - William R Zhang
- Kidney Health Research Collaborative, Department of Medicine, San Francisco Veterans Affairs Medical Center and University of California, 533 Parnassus Avenue, U404, Box 0532, San Francisco, CA, 94143, USA
| | - Simon B Ascher
- Kidney Health Research Collaborative, Department of Medicine, San Francisco Veterans Affairs Medical Center and University of California, 533 Parnassus Avenue, U404, Box 0532, San Francisco, CA, 94143, USA.,Department of Medicine, University of California, Los Angeles, CA, USA
| | - Vasantha Jotwani
- Kidney Health Research Collaborative, Department of Medicine, San Francisco Veterans Affairs Medical Center and University of California, 533 Parnassus Avenue, U404, Box 0532, San Francisco, CA, 94143, USA
| | - Carl Grunfeld
- Kidney Health Research Collaborative, Department of Medicine, San Francisco Veterans Affairs Medical Center and University of California, 533 Parnassus Avenue, U404, Box 0532, San Francisco, CA, 94143, USA
| | - Chirag R Parikh
- Department of Medicine, Section of Nephrology, Yale University, New Haven, CT, USA
| | - Derek Ng
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Frank J Palella
- Division of Infectious Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ken Ho
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Seble Kassaye
- Georgetown University Medical Center, Washington, DC, USA
| | - Anjali Sharma
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Mardge Cohen
- Department of Medicine, Stroger Hospital and Rush University, Chicago, IL, USA
| | - Ruibin Wang
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michelle M Estrella
- Kidney Health Research Collaborative, Department of Medicine, San Francisco Veterans Affairs Medical Center and University of California, 533 Parnassus Avenue, U404, Box 0532, San Francisco, CA, 94143, USA
| |
Collapse
|
38
|
Role of Epidermal Growth Factor Receptor (EGFR) and Its Ligands in Kidney Inflammation and Damage. Mediators Inflamm 2018; 2018:8739473. [PMID: 30670929 PMCID: PMC6323488 DOI: 10.1155/2018/8739473] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/29/2018] [Accepted: 11/07/2018] [Indexed: 12/29/2022] Open
Abstract
Chronic kidney disease (CKD) is characterized by persistent inflammation and progressive fibrosis, ultimately leading to end-stage renal disease. Although many studies have investigated the factors involved in the progressive deterioration of renal function, current therapeutic strategies only delay disease progression, leaving an unmet need for effective therapeutic interventions that target the cause behind the inflammatory process and could slow down or reverse the development and progression of CKD. Epidermal growth factor receptor (EGFR) (ERBB1), a membrane tyrosine kinase receptor expressed in the kidney, is activated after renal damage, and preclinical studies have evidenced its potential as a therapeutic target in CKD therapy. To date, seven official EGFR ligands have been described, including epidermal growth factor (EGF) (canonical ligand), transforming growth factor-α, heparin-binding epidermal growth factor, amphiregulin, betacellulin, epiregulin, and epigen. Recently, the connective tissue growth factor (CTGF/CCN2) has been described as a novel EGFR ligand. The direct activation of EGFR by its ligands can exert different cellular responses, depending on the specific ligand, tissue, and pathological condition. Among all EGFR ligands, CTGF/CCN2 is of special relevance in CKD. This growth factor, by binding to EGFR and downstream signaling pathway activation, regulates renal inflammation, cell growth, and fibrosis. EGFR can also be “transactivated” by extracellular stimuli, including several key factors involved in renal disease, such as angiotensin II, transforming growth factor beta (TGFB), and other cytokines, including members of the tumor necrosis factor superfamily, showing another important mechanism involved in renal pathology. The aim of this review is to summarize the contribution of EGFR pathway activation in experimental kidney damage, with special attention to the regulation of the inflammatory response and the role of some EGFR ligands in this process. Better insights in EGFR signaling in renal disease could improve our current knowledge of renal pathology contributing to therapeutic strategies for CKD development and progression.
Collapse
|
39
|
Latcha S, Jaimes EA, Gutgarts V, Seshan S. Case of Proteinuria, Worsening Hypertension, and Glomerular Endotheliosis With Erlotinib and Gefitinib. Kidney Int Rep 2018; 3:1477-1481. [PMID: 30450475 PMCID: PMC6224627 DOI: 10.1016/j.ekir.2018.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Affiliation(s)
- Sheron Latcha
- Renal Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pathology, Weill Cornell Medical College, New York, New York, USA
| | - Edgar A Jaimes
- Renal Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Victoria Gutgarts
- Renal Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA.,Division of Pathology, Weill Cornell Medical College, New York, New York, USA
| | - Surya Seshan
- Division of Pathology, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
40
|
Ke B, Chen Y, Tu W, Ye T, Fang X, Yang L. Inhibition of HDAC6 activity in kidney diseases: a new perspective. Mol Med 2018; 24:33. [PMID: 30134806 PMCID: PMC6019784 DOI: 10.1186/s10020-018-0027-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 05/16/2018] [Indexed: 12/18/2022] Open
Abstract
Histone deacetylase 6 (HDAC6), a cytoplasmic enzyme that plays important roles in many biological processes, is one isoform of a family of HDAC enzymes that catalyse the removal of functional acetyl groups from proteins. HDAC6 stands out from the other members of this family because it almost exclusively deacetylates cytoplasmic proteins and exerts deacetylation-independent effects, which has led to the successful development of relatively isoform-specific inhibitors of its enzymatic action. Numerous studies have recently demonstrated that HDAC6 expression and activity are increased in kidney disease, such as autosomal dominant polycystic kidney disease (ADPKD), renal fibrosis, and acute kidney injury (AKI), among others. Moreover, HDAC6 inhibitors have been investigated for use in treating these diseases. In fact, HDAC6 inhibitors effectively limit the progression of kidney diseases, suggesting that targeting HDAC6 may provide a novel treatment approach. However, the primary challenge in developing HDAC6-targeted therapies is understanding how the renoprotective effect of NDAC6 inhibitors can be selectively harnessed. Here, we discuss the unique function of HDAC6 and recapitulate the alluring potential of its inhibitors in kidney diseases.
Collapse
Affiliation(s)
- Ben Ke
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yanxia Chen
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wei Tu
- Department of Endocrinology, The Affiliated Tongji Hospital of Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Ting Ye
- Department of Intensive Care Unit, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| | - Xiangdong Fang
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China. .,, Nanchang, People's Republic of China.
| | - Liping Yang
- Department of Breast, Jiangxi Cancer Hospital, Nanchang, 330006, Jiangxi, China. .,, Nanchang, People's Republic of China.
| |
Collapse
|
41
|
Sjaarda J, Gerstein HC, Yusuf S, Treleaven D, Walsh M, Mann JFE, Hess S, Paré G. Blood HER2 and Uromodulin as Causal Mediators of CKD. J Am Soc Nephrol 2018; 29:1326-1335. [PMID: 29511113 DOI: 10.1681/asn.2017070812] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 01/18/2018] [Indexed: 01/04/2023] Open
Abstract
Many biomarkers have been epidemiologically linked with CKD; however, the possibility that such associations are due to reverse causation or confounding limits the utility of these biomarkers. To overcome this limitation, we used a Mendelian randomization (MR) approach to identify causal mediators of CKD. We performed MR by first identifying genetic determinants of 227 serum protein biomarkers assayed in 4147 participants of the Outcome Reduction with Initial Glargine Intervention (ORIGIN) trial who had early or prediabetes, and assessing the effects of these biomarkers on CKD in the CKD genetics consortium (n=117,165; 12,385 cases) using the inverse-variance weighted (fixed-effects) method. We then estimated the relationship between the serum concentration of each biomarker identified and incident CKD in ORIGIN participants. MR identified uromodulin (UMOD) and human EGF receptor 2 (HER2) as novel, causal mediators of CKD (UMOD: odds ratio [OR], 1.30 per SD; 95% confidence interval [95% CI], 1.25 to 1.35; P<5×10-20; HER2: OR, 1.30 per SD; 95% CI, 1.14 to 1.48; P=8.0×10-5). Consistent with these findings, blood HER2 concentration associated with CKD events in ORIGIN participants (OR, 1.07 per SD; 95% CI, 1.01 to 1.13; P=0.01). Additional exploratory MR analyses identified angiotensin-converting enzyme (ACE) as a regulator of HER2 levels (β=0.13 per SD; 95% CI, 0.08 to 0.16; P=2.5×10-7). This finding was corroborated by an inverse relationship between ACE inhibitor use and HER2 levels. Thus, UMOD and HER2 are independent causal mediators of CKD in humans, and serum HER2 levels are regulated in part by ACE. These biomarkers are potential therapeutic targets for CKD prevention.
Collapse
Affiliation(s)
- Jennifer Sjaarda
- Population Health Research Institute and.,Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada.,Departments of Medical Sciences
| | - Hertzel C Gerstein
- Population Health Research Institute and.,Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada
| | | | | | - Michael Walsh
- Population Health Research Institute and.,Medicine.,Health Research Methods, Evaluation and Impact, and
| | - Johannes F E Mann
- Department of Nephrology and Hypertension, University Hospital, Friedrich-Alexander University, Erlangen-Nuremberg, Germany.,Department of Medicine IV, University of Erlangen-Nurnberg, Erlangen, Germany
| | - Sibylle Hess
- Sanofi Aventis Deutschland GmbH, Research and Development Division, Translational Medicine and Early Development, Biomarkers and Clinical Bioanalyses, Frankfurt, Germany; and
| | - Guillaume Paré
- Population Health Research Institute and .,Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Michael G. DeGroote School of Medicine, Hamilton, Ontario, Canada.,Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
42
|
Sloan-Lancaster J, Raddad E, Deeg MA, Eli M, Flynt A, Tumlin J. Evaluation of the Safety, Pharmacokinetics, Pharmacodynamics, and Efficacy After Single and Multiple Dosings of LY3016859 in Healthy Subjects and Patients With Diabetic Nephropathy. Clin Pharmacol Drug Dev 2018; 7:759-772. [PMID: 29385323 DOI: 10.1002/cpdd.436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 12/07/2017] [Indexed: 11/06/2022]
Abstract
Two phase 1 studies (TGAA and TGAB) evaluated the safety, pharmacokinetics, pharmacodynamics, and efficacy of LY3016859 (LY), a monoclonal antibody that binds epiregulin and transforming growth factor α (TGF-α), administered intravenously or subcutaneously. In TGAA, 56 healthy subjects received a single dose of LY (0.1-750 mg intravenously, 50 mg subcutaneously) or placebo. In TGAB part A, 15 patients with diabetic nephropathy (DN) received 2 doses of LY (10-750 mg intravenously) or placebo, and in TGAB part B, 45 patients with DN received 5 doses of LY (50-750 mg intravenously) or placebo. Pharmacokinetics, pharmacodynamics, anti-LY antibodies, and change in proteinuria and albuminuria were evaluated. Single and multiple doses of LY administered 3 weeks apart were well tolerated. Pharmacokinetics were nonlinear in healthy subjects and patients with DN, indicating target-mediated drug disposition. Epiregulin level increased in both studies, and TGF-α levels increased in the TGAB study, consistent with target engagement; however, LY treatment did not significantly reduce proteinuria or albuminuria in patients with DN. There was no obvious effect of LY on the disease-related biomarkers monocyte chemoattractant protein-1, synaptopodin, or transferrin. Although LY administration resulted in a high frequency of anti-LY antibodies, pharmacokinetics, target engagement, and efficacy were not impacted.
Collapse
Affiliation(s)
| | - Eyas Raddad
- Chorus, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Mark A Deeg
- Chorus, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Michelle Eli
- Chorus, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - Amy Flynt
- PharPoint Research, Inc., Durham, NC, USA
| | | |
Collapse
|
43
|
A Two-Stage Whole-Genome Gene Expression Association Study of Young-Onset Hypertension in Han Chinese Population of Taiwan. Sci Rep 2018; 8:1800. [PMID: 29379041 PMCID: PMC5789005 DOI: 10.1038/s41598-018-19520-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 01/03/2018] [Indexed: 12/31/2022] Open
Abstract
Hypertension is an important public health problem in the world. Since the intermediate position of the gene expression between genotype and phenotype makes it suitable to link genotype to phenotype, we carried out a two-stage whole-genome gene expression association study to find differentially expressed genes and pathways for hypertension. In the first stage, 126 cases and 149 controls were used to find out the differentially expressed genes. In the second stage, an independent set of samples (127 cases and 150 controls) was used to validate the results. Additionally, we conducted a gene set enrichment analysis (GSEA) to search for differentially affected pathways. A total of nine genes were implicated in the first stage (Bonferroni-corrected p-value < 0.05). Among these genes, ZRANB1, FAM110A, PREP, ANKRD9 and LAMB2 were also differentially expressed in an existing database of hypertensive mouse model (GSE19817). A total of 16 pathways were identified by the GSEA. ZRANB1 and six pathways identified are related to TNF-α. Three pathways are related to interleukin, one to metabolic syndrome, and one to Hedgehog signaling. Identification of these genes and pathways suggest the importance of 1. inflammation, 2. visceral fat metabolism, and 3. adipocytes and osteocytes homeostasis in hypertension mechanisms and complications.
Collapse
|
44
|
Lai X, Zhong L, Fu HX, Dang S, Wang X, Zhang N, Feng GK, Liu ZQ, Wang X, Wang L. Effects of neuregulin-1 on autonomic nervous system remodeling post-myocardial infarction in a rat model. Neural Regen Res 2017; 12:1905-1910. [PMID: 29239338 PMCID: PMC5745846 DOI: 10.4103/1673-5374.219054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Sympathetic nerve and vagus nerve remodeling play an important part in cardiac function post-myocardial infarction (MI). Increasing evidence indicates that neuregulin-1 (NRG-1) improves cardiac function following heart failure. Since its impact on cardiac function and neural remodeling post-MI is poorly understood, we aimed to investigate the role of NRG-1 in autonomic nervous system remodeling post-MI. Forty-five Sprague-Dawley rats were equally randomized into three groups: sham (with the left anterior descending coronary artery exposed but without ligation), MI (left anterior descending coronary artery ligation), and MI plus NRG-1 (left anterior descending coronary artery ligation followed by intraperitoneal injection of NRG-1 (10 μg/kg, once daily for 7 days)). At 4 weeks after MI, echocardiography was used to detect the rat cardiac function by measuring the left ventricular end-systolic inner diameter, left ventricular diastolic diameter, left ventricular end-systolic volume, left ventricular end-diastolic volume, left ventricular ejection fraction, and left ventricular fractional shortening. mRNA and protein expression levels of tyrosine hydroxylase, growth associated protein-43 (neuronal specific protein), nerve growth factor, choline acetyltransferase (vagus nerve marker), and vesicular acetylcholine transporter (cardiac vagal nerve fiber marker) in ischemic myocardia were detected by real-time PCR and western blot assay to assess autonomous nervous remodeling. After MI, the rat cardiac function deteriorated significantly, and it was significantly improved after NRG-1 injection. Compared with the MI group, mRNA and protein levels of tyrosine hydroxylase and growth associated protein-43, as well as choline acetyltransferase mRNA level significantly decreased in the MI plus NRG-1 group, while mRNA and protein levels of nerve growth factor and vesicular acetylcholine transporters, as well as choline acetyltransferase protein level slightly decreased. Our results indicate that NRG-1 can improve cardiac function and regulate sympathetic and vagus nerve remodeling post-MI, thus reaching a new balance of the autonomic nervous system to protect the heart from injury.
Collapse
Affiliation(s)
- Xin Lai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei Province; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province, China
| | - Liang Zhong
- Wuhan Medical & Healthcare Center for Women and Children, Wuhan, Hubei Province, China
| | - Hai-Xia Fu
- Department of Cardiology, Henan Province People's Hospital, Zhengzhou, Henan Province, China
| | - Song Dang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei Province; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province, China
| | - Xin Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei Province; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province, China
| | - Ning Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei Province; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province, China
| | - Gao-Ke Feng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei Province; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province, China
| | - Zi-Qiang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei Province; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province, China
| | - Xi Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province; Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei Province; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province, China
| | - Long Wang
- Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei Province; Hubei Key Laboratory of Cardiology, Wuhan, Hubei Province, China
| |
Collapse
|
45
|
Mahalingam SM, Dudkin V, Goldberg S, Klein D, Yi F, Singhal S, O’Neil KT, Low PS. Evaluation of a Centyrin-Based Near-Infrared Probe for Fluorescence-Guided Surgery of Epidermal Growth Factor Receptor Positive Tumors. Bioconjug Chem 2017; 28:2865-2873. [PMID: 28945346 PMCID: PMC11017363 DOI: 10.1021/acs.bioconjchem.7b00566] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tumor-targeted near-infrared fluorescent dyes have the potential to improve cancer surgery by enabling surgeons to locate and resect more malignant lesions where good visualization tools are required to ensure complete removal of malignant tissue. Although the tumor-targeted fluorescent dyes used in humans to date have been either small organic molecules or high molecular weight antibodies, low molecular weight protein scaffolds have attracted significant attention because they penetrate solid tumors almost as efficiently as small molecules, but can be infinitely mutated to bind almost any antigen. Here we describe the use of a 10 kDa protein scaffold, a Centyrin, to target a near-infrared fluorescent dye to tumors that overexpress the epidermal growth factor receptor (EGFR) for fluorescence-guided surgery (FGS). We have developed and optimized the dose and time required for imaging small tumor burdens with minimal background fluorescence in real-time fluorescence-guided surgery of EGFR-expressing tumor xenografts in murine models. We demonstrate that the Centyrin-near-infrared dye conjugate (CNDC) binds selectively to human EGFR+ cancer cells with an EC50 of 2 nM, localizes to EGFR+ tumor xenografts in athymic nude mice and that uptake of the dye in xenografts is significantly reduced when EGFR are blocked by preinjection of excess unlabeled Centyrin. Taken together, these data suggest that CNDCs can be used for intraoperative identification and surgical removal of EGFR-expressing lesions and that Centyrins targeted to other tumor-specific antigens should prove similarly useful in fluorescence guided surgery of cancer. In addition, we demonstrate that the CNDC is detected in the NIR region of the spectrum and can be utilized for fluorescence-guided surgery (FGS). In addition, we propose that with its eventual complete clearance from EGFR-negative tissues and its quantitative retention in the tumor mass for >24 h, a Centyrin-targeted NIR dye should provide excellent tumor contrast when injected at least 6-8 h before initiation of cancer surgery in human patients.
Collapse
Affiliation(s)
- Sakkarapalayam M. Mahalingam
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Vadim Dudkin
- Janssen Research & Development, 1400 McKean Road, Springhouse PA 19477, United States
| | - Shalom Goldberg
- Janssen Research & Development, 1400 McKean Road, Springhouse PA 19477, United States
| | - Donna Klein
- Janssen Research & Development, 1400 McKean Road, Springhouse PA 19477, United States
| | - Fang Yi
- Janssen Research & Development, 1400 McKean Road, Springhouse PA 19477, United States
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Karyn T. O’Neil
- Janssen Research & Development, 1400 McKean Road, Springhouse PA 19477, United States
| | - Philip S. Low
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
46
|
Zepeda-Orozco D, Wen HM, Hamilton BA, Raikwar NS, Thomas CP. EGF regulation of proximal tubule cell proliferation and VEGF-A secretion. Physiol Rep 2017; 5:5/18/e13453. [PMID: 28963126 PMCID: PMC5617933 DOI: 10.14814/phy2.13453] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 07/31/2017] [Accepted: 08/17/2017] [Indexed: 11/24/2022] Open
Abstract
Proximal tubule cell (PTC) proliferation is critical for tubular regeneration and recovery from acute kidney injury. Epidermal growth factor (EGF) and vascular endothelial growth factor (VEGF‐A) are important for the maintenance of tubulointerstitial integrity and can stimulate PTC proliferation. We utilized HK‐2 cells, an immortalized human PTC line, to characterize the EGF‐dependent regulation of VEGF‐A secretion and proliferation in PTCs. We demonstrate that EGF stimulates VEGF‐A secretion via the EGF receptor (EGFR) and stimulates cell proliferation via activation of the VEGF receptor, VEGFR‐2. EGFR activation promotes MAPK (ERK1/2) activation and HIF‐1α expression, which are required for basal and EGF‐stimulated VEGF‐A secretion. EGF also stimulates the phosphorylation of P70S6 kinase (P70S6K), the downstream target of mTORC1. Rapamycin decreased basal and EGF stimulated HIF‐1α and enhanced MAPK (ERK1/2) activation, while MAPK (ERK/12) inhibition downregulated HIF‐1α expression and the phosphorylation of p70S6K. EGF stimulation of p70S6K was also independent of p‐AKT. Inhibition of the mTORC1 pathway with rapamycin abolished phosphorylation of p70S6K but had no effect on VEGF‐A secretion, indicating that EGF‐stimulated VEGF‐A secretion did not require mTORC1 pathway activation. We demonstrate evidence of a complex crosstalk between the MAPK/ERK and mTORC1 pathways, wherein MAPK (ERK1/2) activation stimulates p‐P70S6K, while p‐P70S6K activation seems to inhibit MAPK (ERK1/2) in EGF‐treated HK‐2 cells. Our results suggest that EGF stimulates MAPK (ERK1/2) in HK‐2 cells, which in turn increases HIF‐1α expression and VEGF‐A secretion, indicating that VEGF‐A mediates EGF‐stimulated cell proliferation as an autocrine proximal tubular epithelial cell growth factor.
Collapse
Affiliation(s)
- Diana Zepeda-Orozco
- Division of Pediatric Nephrology, Stead Family Department of Pediatrics, Dialysis and Transplantation, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Hsiang M Wen
- Division of Pediatric Nephrology, Stead Family Department of Pediatrics, Dialysis and Transplantation, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Bradley A Hamilton
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Nandita S Raikwar
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Christie P Thomas
- Division of Pediatric Nephrology, Stead Family Department of Pediatrics, Dialysis and Transplantation, University of Iowa Carver College of Medicine, Iowa City, Iowa.,Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa.,VA Medical Center, Iowa City, Iowa
| |
Collapse
|
47
|
Caetano-Pinto P, Jamalpoor A, Ham J, Goumenou A, Mommersteeg M, Pijnenburg D, Ruijtenbeek R, Sanchez-Romero N, van Zelst B, Heil SG, Jansen J, Wilmer MJ, van Herpen CML, Masereeuw R. Cetuximab Prevents Methotrexate-Induced Cytotoxicity in Vitro through Epidermal Growth Factor Dependent Regulation of Renal Drug Transporters. Mol Pharm 2017; 14:2147-2157. [PMID: 28493713 PMCID: PMC5462489 DOI: 10.1021/acs.molpharmaceut.7b00308] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
![]()
The combination of methotrexate with
epidermal growth factor receptor
(EGFR) recombinant antibody, cetuximab, is currently being investigated
in treatment of head and neck carcinoma. As methotrexate is cleared
by renal excretion, we studied the effect of cetuximab on renal methotrexate
handling. We used human conditionally immortalized proximal tubule
epithelial cells overexpressing either organic anion transporter 1
or 3 (ciPTEC-OAT1/ciPTEC-OAT3) to examine OAT1 and OAT3, and the efflux
pumps breast cancer resistance protein (BCRP), multidrug resistance
protein 4 (MRP4), and P-glycoprotein (P-gp) in methotrexate handling
upon EGF or cetuximab treatment. Protein kinase microarrays and knowledge-based
pathway analysis were used to predict EGFR-mediated transporter regulation.
Cytotoxic effects of methotrexate were evaluated using the dimethylthiazol
bromide (MTT) viability assay. Methotrexate inhibited OAT-mediated
fluorescein uptake and decreased efflux of Hoechst33342 and glutathione-methylfluorescein
(GS-MF), which suggested involvement of OAT1/3, BCRP, and MRP4 in
transepithelial transport, respectively. Cetuximab reversed the EGF-increased
expression of OAT1 and BCRP as well as their membrane expressions
and transport activities, while MRP4 and P-gp were increased. Pathway
analysis predicted cetuximab-induced modulation of PKC and PI3K pathways
downstream EGFR/ERBB2/PLCg. Pharmacological inhibition of ERK decreased
expression of OAT1 and BCRP, while P-gp and MRP4 were increased. AKT
inhibition reduced all transporters. Exposure to methotrexate for
24 h led to a decreased viability, an effect that was reversed by
cetuximab. In conclusion, cetuximab downregulates OAT1 and BCRP while
upregulating P-gp and MRP4 through an EGFR-mediated regulation of
PI3K-AKT and MAPKK-ERK pathways. Consequently, cetuximab attenuates
methotrexate-induced cytotoxicity, which opens possibilities for further
research into nephroprotective comedication therapies.
Collapse
Affiliation(s)
- Pedro Caetano-Pinto
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , 3584 CG Utrecht, The Netherlands
| | - Amer Jamalpoor
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , 3584 CG Utrecht, The Netherlands
| | - Janneke Ham
- Department of Oncology, Radboud University Medical Center , 6525 GA Nijmegen, The Netherlands
| | - Anastasia Goumenou
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , 3584 CG Utrecht, The Netherlands
| | | | | | | | - Natalia Sanchez-Romero
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , 3584 CG Utrecht, The Netherlands.,Centro Investigación Biomédica de Aragón (CIBA), 50009 Zaragoza, Spain
| | - Bertrand van Zelst
- Department of Clinical Chemistry, ErasmusMC , 3015 CE Rotterdam, The Netherlands
| | - Sandra G Heil
- Department of Clinical Chemistry, ErasmusMC , 3015 CE Rotterdam, The Netherlands
| | - Jitske Jansen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , 3584 CG Utrecht, The Netherlands
| | - Martijn J Wilmer
- Department of Pharmacology and Toxicology, Radboud Institute of Molecular Life Sciences, Radboudumc , 6500 HB Nijmegen, The Netherlands
| | - Carla M L van Herpen
- Department of Oncology, Radboud University Medical Center , 6525 GA Nijmegen, The Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University , 3584 CG Utrecht, The Netherlands
| |
Collapse
|
48
|
Gade AM, Meadows MK, Ellington AD, Anslyn EV. Differential array sensing for cancer cell classification and novelty detection. Org Biomol Chem 2017; 15:9866-9874. [DOI: 10.1039/c7ob02174g] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A series of semi-specific peptides reported in the literature to bind various epitopes on cell surfaces were used in a differential sensing array to pattern cell line identity.
Collapse
Affiliation(s)
| | | | - Andrew D. Ellington
- Institute for Cellular and Molecular Biology
- The University of Texas at Austin
- Austin
- USA
| | - Eric V. Anslyn
- Department of Chemistry
- The University of Texas at Austin
- Austin
- USA
| |
Collapse
|
49
|
Shi X, Xu Y, Zhang C, Feng L, Sun Z, Han J, Su F, Zhang Y, Li C, Li X. Subpathway-LNCE: Identify dysfunctional subpathways competitively regulated by lncRNAs through integrating lncRNA-mRNA expression profile and pathway topologies. Oncotarget 2016; 7:69857-69870. [PMID: 27634882 PMCID: PMC5342520 DOI: 10.18632/oncotarget.12005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 09/02/2016] [Indexed: 12/14/2022] Open
Abstract
Recently, studies have reported that long noncoding RNAs (lncRNAs) can act as modulators of mRNAs through competitively binding to microRNAs (miRNAs) and have relevance to tumorigenesis as well as other diseases. Identify lncRNA competitively regulated subpathway not only can gain insight into the initiation and progression of disease, but also help for understanding the functional roles of lncRNAs in the disease context. Here, we present an effective method, Subpathway-LNCE, which was specifically designed to identify lncRNAs competitively regulated functions and the functional roles of these competitive regulation lncRNAs have not be well characterized in diseases. Moreover, the method integrated lncRNA-mRNA expression profile and pathway topologies. Using prostate cancer datasets and LUAD data sets, we confirmed the effectiveness of our method in identifying disease associated dysfunctional subpathway that regulated by lncRNAs. By analyzing kidney renal clear cell carcinoma related lncRNA competitively regulated subpathway network, we show that Subpathway-LNCE can help uncover disease key lncRNAs. Furthermore, we demonstrated that our method is reproducible and robust. Subpathway-LNCE provide a flexible tool to identify lncRNA competitively regulated signal subpathways underlying certain condition, and help to expound the functional roles of lncRNAs in various status. Subpathway-LNCE has been developed as an R package freely available at https://cran.rstudio.com/web/packages/SubpathwayLNCE/.
Collapse
Affiliation(s)
- Xinrui Shi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yanjun Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Chunlong Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Li Feng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Zeguo Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Junwei Han
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Fei Su
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Chunquan Li
- Department of Medical Informatics, Daqing Campus, Harbin Medical University, Daqing, 163319, China
| | - Xia Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
50
|
Cheung PW, Nomura N, Nair AV, Pathomthongtaweechai N, Ueberdiek L, Lu HAJ, Brown D, Bouley R. EGF Receptor Inhibition by Erlotinib Increases Aquaporin 2-Mediated Renal Water Reabsorption. J Am Soc Nephrol 2016; 27:3105-3116. [PMID: 27694161 PMCID: PMC5042667 DOI: 10.1681/asn.2015080903] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/28/2016] [Indexed: 01/04/2023] Open
Abstract
Nephrogenic diabetes insipidus (NDI) is caused by impairment of vasopressin (VP) receptor type 2 signaling. Because potential therapies for NDI that target the canonical VP/cAMP/protein kinase A pathway have so far proven ineffective, alternative strategies for modulating aquaporin 2 (AQP2) trafficking have been sought. Successful identification of compounds by our high-throughput chemical screening assay prompted us to determine whether EGF receptor (EGFR) inhibitors stimulate AQP2 trafficking and reduce urine output. Erlotinib, a selective EGFR inhibitor, enhanced AQP2 apical membrane expression in collecting duct principal cells and reduced urine volume by 45% after 5 days of treatment in mice with lithium-induced NDI. Similar to VP, erlotinib increased exocytosis and decreased endocytosis in LLC-PK1 cells, resulting in a significant increase in AQP2 membrane accumulation. Erlotinib increased phosphorylation of AQP2 at Ser-256 and Ser-269 and decreased phosphorylation at Ser-261 in a dose-dependent manner. However, unlike VP, the effect of erlotinib was independent of cAMP, cGMP, and protein kinase A. Conversely, EGF reduced VP-induced AQP2 Ser-256 phosphorylation, suggesting crosstalk between VP and EGF in AQP2 trafficking and a role of EGF in water homeostasis. These results reveal a novel pathway that contributes to the regulation of AQP2-mediated water reabsorption and suggest new potential therapeutic strategies for NDI treatment.
Collapse
Affiliation(s)
- Pui W Cheung
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Naohiro Nomura
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Anil V Nair
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Nutthapoom Pathomthongtaweechai
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lars Ueberdiek
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Hua A Jenny Lu
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dennis Brown
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Richard Bouley
- Center for Systems Biology, Program in Membrane Biology, Division of Nephrology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|