1
|
Götzinger F, Kunz M, Lauder L, Böhm M, Mahfoud F. New ways of mitigating aldosterone in cardiorenal disease. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2024; 10:557-565. [PMID: 38986505 DOI: 10.1093/ehjcvp/pvae049] [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: 05/08/2024] [Revised: 06/11/2024] [Accepted: 07/09/2024] [Indexed: 07/12/2024]
Abstract
Steroidal mineralocorticoid receptor antagonists (MRAs) bind to the mineralocorticoid receptor and antagonize the effects of aldosterone, which contributes to the development and progression of cardio- and renovascular diseases. Guidelines recommend steroidal MRAs in patients with heart failure with reduced or mildly reduced ejection fraction, as they reduce morbidity and mortality. In heart failure with preserved ejection fraction, MRAs have not convincingly shown to improve prognosis. Steroidal MRAs delay the progression of chronic kidney disease, reduce proteinuria and lower blood pressure in resistant hypertension but can induce hyperkalaemia. Due to their limited selectivity to the mineralocorticoid receptor, steroidal MRAs can cause significant adverse effects, i.e. libido loss, erectile dysfunction, gynaecomastia, and amenorrhoea, leading to low rates of persistance. Against this background, new avenues for developing non-steroidal, selective (ns)MRAs and aldosterone-synthase inhibitors have been taken. Finerenone has been shown to delay the progression of diabetic nephropathy and lower the incidence of heart failure hospitalizations in patients with chronic kidney disease and diabetes compared with placebo. Finerenone has therefore been recommended by the 2023 European Society of Cardiology Guidelines for the management of diabetes in patients with type 2 diabetes and chronic kidney disease. Further randomized controlled trials assessing the safety and effectiveness of finerenone in patients with heart failure are currently ongoing. Esaxerenone provides antihypertensive effects and has been approved for the treatment of hypertension in Japan. Baxdrostat and lorundostat, novel selective aldosterone-synthase inhibitors, are currently under investigation. In phase II trials, baxdrostat and lorundostat were safe and effective in lowering blood pressure in resistant hypertension. In this review, we summarize and critically discuss the evidence for new drugs mitigating aldosterone in heart failure, hypertension, and chronic kidney disease.
Collapse
Affiliation(s)
- Felix Götzinger
- Department of Internal Medicine III-Cardiology, Angiology and Intensive Care Medicine, Homburg University Hospital, Saarland University, Kirrberger Str. 100, Homburg 66424, Germany
- Department of Cardiology, University Heart Center Basel, University Hospital Basel, Am Petersgraben 4, Basel 4031, Switzerland
| | - Michael Kunz
- Department of Internal Medicine III-Cardiology, Angiology and Intensive Care Medicine, Homburg University Hospital, Saarland University, Kirrberger Str. 100, Homburg 66424, Germany
- Department of Cardiology, University Heart Center Basel, University Hospital Basel, Am Petersgraben 4, Basel 4031, Switzerland
| | - Lucas Lauder
- Department of Internal Medicine III-Cardiology, Angiology and Intensive Care Medicine, Homburg University Hospital, Saarland University, Kirrberger Str. 100, Homburg 66424, Germany
- Department of Cardiology, University Heart Center Basel, University Hospital Basel, Am Petersgraben 4, Basel 4031, Switzerland
| | - Michael Böhm
- Department of Internal Medicine III-Cardiology, Angiology and Intensive Care Medicine, Homburg University Hospital, Saarland University, Kirrberger Str. 100, Homburg 66424, Germany
| | - Felix Mahfoud
- Department of Internal Medicine III-Cardiology, Angiology and Intensive Care Medicine, Homburg University Hospital, Saarland University, Kirrberger Str. 100, Homburg 66424, Germany
- Department of Cardiology, University Heart Center Basel, University Hospital Basel, Am Petersgraben 4, Basel 4031, Switzerland
| |
Collapse
|
2
|
Niculae A, Gherghina ME, Peride I, Tiglis M, Nechita AM, Checherita IA. Pathway from Acute Kidney Injury to Chronic Kidney Disease: Molecules Involved in Renal Fibrosis. Int J Mol Sci 2023; 24:14019. [PMID: 37762322 PMCID: PMC10531003 DOI: 10.3390/ijms241814019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/30/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Acute kidney injury (AKI) is one of the main conditions responsible for chronic kidney disease (CKD), including end-stage renal disease (ESRD) as a long-term complication. Besides short-term complications, such as electrolyte and acid-base disorders, fluid overload, bleeding complications or immune dysfunctions, AKI can develop chronic injuries and subsequent CKD through renal fibrosis pathways. Kidney fibrosis is a pathological process defined by excessive extracellular matrix (ECM) deposition, evidenced in chronic kidney injuries with maladaptive architecture restoration. So far, cited maladaptive kidney processes responsible for AKI to CKD transition were epithelial, endothelial, pericyte, macrophage and fibroblast transition to myofibroblasts. These are responsible for smooth muscle actin (SMA) synthesis and abnormal renal architecture. Recently, AKI progress to CKD or ESRD gained a lot of interest, with impressive progression in discovering the mechanisms involved in renal fibrosis, including cellular and molecular pathways. Risk factors mentioned in AKI progression to CKD are frequency and severity of kidney injury, chronic diseases such as uncontrolled hypertension, diabetes mellitus, obesity and unmodifiable risk factors (i.e., genetics, older age or gender). To provide a better understanding of AKI transition to CKD, we have selected relevant and updated information regarding the risk factors responsible for AKIs unfavorable long-term evolution and mechanisms incriminated in the progression to a chronic state, along with possible therapeutic approaches in preventing or delaying CKD from AKI.
Collapse
Affiliation(s)
- Andrei Niculae
- Department of Nephrology, Clinical Department No. 3, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Mihai-Emil Gherghina
- Department of Nephrology, Ilfov County Emergency Clinical Hospital, 022104 Bucharest, Romania
| | - Ileana Peride
- Department of Nephrology, Clinical Department No. 3, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Mirela Tiglis
- Department of Anesthesia and Intensive Care, Emergency Clinical Hospital of Bucharest, 014461 Bucharest, Romania
| | - Ana-Maria Nechita
- Department of Nephrology, “St. John” Emergency Clinical Hospital, 042122 Bucharest, Romania
| | | |
Collapse
|
3
|
Abstract
Kidney disease is associated with adverse consequences in many organs beyond the kidney, including the heart, lungs, brain, and intestines. The kidney-intestinal cross talk involves intestinal epithelial damage, dysbiosis, and generation of uremic toxins. Recent studies reveal that kidney injury expands the intestinal lymphatics, increases lymphatic flow, and alters the composition of mesenteric lymph. The intestinal lymphatics, like blood vessels, are a route for transporting potentially harmful substances generated by the intestines. The lymphatic architecture and actions are uniquely suited to take up and transport large macromolecules, functions that differentiate them from blood vessels, allowing them to play a distinct role in a variety of physiological and pathological processes. Here, we focus on the mechanisms by which kidney diseases result in deleterious changes in intestinal lymphatics and consider a novel paradigm of a vicious cycle of detrimental organ cross talk. This concept involves kidney injury-induced modulation of intestinal lymphatics that promotes production and distribution of harmful factors, which in turn contributes to disease progression in distant organ systems.
Collapse
Affiliation(s)
- Jianyong Zhong
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and Immunology (J.Z., H.-C.Y., A.B.F.), Vanderbilt University Medical Center, Nashville, TN
| | - Annet Kirabo
- Department of Molecular Physiology and Biophysics (A.K.), Vanderbilt University Medical Center, Nashville, TN
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN (A.K.)
| | - Hai-Chun Yang
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and Immunology (J.Z., H.-C.Y., A.B.F.), Vanderbilt University Medical Center, Nashville, TN
| | - Agnes B Fogo
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
- Department of Pathology, Microbiology and Immunology (J.Z., H.-C.Y., A.B.F.), Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine (A.B.F.), Vanderbilt University Medical Center, Nashville, TN
| | - Elaine L Shelton
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Valentina Kon
- Department of Pediatrics (J.Z., H.-C.Y., A.B.F., E.L.S., V.K.), Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
4
|
Zhu Y, Yan W, Xu S, Yu X, Sun S, Zhang S, Zhao R, Tao J, Li Y, Li C. Identification of an unrecognized circRNA associated with development of renal fibrosis. Front Genet 2023; 13:964840. [PMID: 36685959 PMCID: PMC9845265 DOI: 10.3389/fgene.2022.964840] [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: 06/09/2022] [Accepted: 12/01/2022] [Indexed: 01/05/2023] Open
Abstract
Backgroud: Renal fibrosis is the common characteristic of chronic kidney disease. Circular RNA plays an essential role in the occurrence and development of Renal fibrosis, but its regulative mechanism remains elusive. Methods: The animal and cell model of Renal fibrosis was established, and RNA-sequencing and real-time polymerase chain reaction (qRT-PCR) experiments were implemented. Subsequently, experiments for detecting apoptosis and proliferation of cell, were carried out, and the isobaric tags for relative and absolute quantification proteomics analyses were performed accordingly. Results: It was found that a newly discovered Circular RNA (circRNA_0002158), is highly expressed in kidneys or cells with fibrosis, implying that this Circular RNA might be associated with the occurrence and development of Renal fibrosis. Subsequently, the overexpression and knockdown of circRNA_0002158 were conducted in the human kidney epithelial cell line (HK-2) cells, and the results indicated that the circRNA_0002158 could inhibit apoptosis, and promote proliferation of cells. The kidney injury-related factors, including Fibronectin and plasminogen activator inhibitor-1 (PAI-1), were decreased in HK-2 cells with overexpression of circRNA_0002158, while the results were reversed in cells with knockdown of circRNA_0002158. Finally, to explore the regulative mechanism of circRNA_0002158, the iTRAQ proteomics analyses were implemented for the cell samples with OE of circRNA_0002158 and its control, it showed that multiple genes and functional pathways were associated with the occurrence and development of Renal fibrosis. Conclusion: CircRNA_0002158 is associated with regulating Renal fibrosis, and may contribute to ameliorating the progression of Renal fibrosis in the future.
Collapse
Affiliation(s)
- Yun Zhu
- Department of Dermatology, The People’s Hospital of Yuxi City, Yuxi, China
| | - Weimin Yan
- Department of Dermatology, The People’s Hospital of Yuxi City, Yuxi, China
| | - Shuangyan Xu
- Department of Dermatology, The People’s Hospital of Yuxi City, Yuxi, China
| | - Xiaochao Yu
- Graduate School, Kunming Medical University, Kunming, China
| | - Shuo Sun
- Graduate School, Kunming Medical University, Kunming, China
| | | | - Ran Zhao
- Graduate School, Kunming Medical University, Kunming, China
| | - Jiayue Tao
- Graduate School, Kunming Medical University, Kunming, China
| | - Yunwei Li
- Department of Urology, The Third Hospital of Shandong Province, Jinan, China,*Correspondence: Yunwei Li, ; Cuie Li,
| | - Cuie Li
- Department of Geriatrics, The People’s Hospital of Yuxi City, Yuxi, China,*Correspondence: Yunwei Li, ; Cuie Li,
| |
Collapse
|
5
|
Tanaka K, Harada H, Kamuro H, Sakai H, Yamamoto A, Tomimatsu M, Ikeda A, Chosokabe R, Tanaka S, Okada Y, Fujio Y, Obana M. Arid5a/IL-6/PAI-1 Signaling Is Involved in the Pathogenesis of Lipopolysaccharide-Induced Kidney Injury. Biol Pharm Bull 2023; 46:1753-1760. [PMID: 38044094 DOI: 10.1248/bpb.b23-00482] [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] [Indexed: 12/05/2023]
Abstract
A systemic inflammatory response leads to widespread organ dysfunction, such as kidney dysfunction. Plasminogen activator inhibitor-1 (PAI-1) is involved in the pathogenesis of inflammatory kidney injury; however, the regulatory mechanism of PAI-1 in injured kidneys remains unclear. PAI-1 is induced by interleukin (IL)-6 in patients with sepsis. In addition, the stabilization of IL-6 is regulated by the adenine-thymine-rich interactive domain-containing protein 5a (Arid5a). Therefore, the aim of the present study was to examine the involvement of Arid5a/IL-6/PAI-1 signaling in lipopolysaccharide (LPS)-induced inflammatory kidney injury. LPS treatment to C57BL/6J mice upregulated Pai-1 mRNA in the kidneys. Enzyme-linked immunosorbent assay (ELISA) revealed that PAI-1 expression was induced in the culture supernatants of LPS-treated human umbilical vein endothelial cells, but not in those of LPS-treated human kidney 2 (HK-2) cells, a tubular cell line. Combined with single-cell analysis, endothelial cells were found to be responsible for PAI-1 elevation in LPS-treated kidneys. Administration of TM5441, a PAI-1 inhibitor, reduced the urinary albumin/creatinine ratio, concomitant with downregulation of Il-6 and Arid5a mRNA expressions. IL-6 treatment in LPS model mice further upregulated Pai-1 mRNA expression compared with LPS alone, accompanied by renal impairment. Furthermore, the expression of Il-6 and Pai-1 mRNA was lower in Arid5a knockout mice than in wild-type mice after LPS treatment. Taken together, the vicious cycle of Arid5a/IL-6/PAI-1 signaling is involved in LPS-induced kidney injury.
Collapse
Affiliation(s)
- Koki Tanaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Hiroki Harada
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Hiroyasu Kamuro
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Hibiki Sakai
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Ayaha Yamamoto
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Masashi Tomimatsu
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Akari Ikeda
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Renya Chosokabe
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Shota Tanaka
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
| | - Yoshiaki Okada
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
- Center for Infectious Disease Education and Research (CiDER), Osaka University
| | - Yasushi Fujio
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
- Center for Infectious Disease Education and Research (CiDER), Osaka University
- Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University
| | - Masanori Obana
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University
- Center for Infectious Disease Education and Research (CiDER), Osaka University
- Institute for Open and Transdisciplinary Research Initiatives (OTRI), Osaka University
- Global Center for Medical Engineering and Informatics (MEI), Osaka University
- Radioisotope Research Center, Institute for Radiation Sciences, Osaka University
| |
Collapse
|
6
|
Dwivedi N, Jamadar A, Mathew S, Fields TA, Rao R. Myofibroblast depletion reduces kidney cyst growth and fibrosis in autosomal dominant polycystic kidney disease. Kidney Int 2023; 103:144-155. [PMID: 36273656 PMCID: PMC9822873 DOI: 10.1016/j.kint.2022.08.036] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 08/10/2022] [Accepted: 08/19/2022] [Indexed: 11/06/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) involves the development and persistent growth of fluid filled kidney cysts. In a recent study, we showed that ADPKD kidney cyst epithelial cells can stimulate the proliferation and differentiation of peri-cystic myofibroblasts. Although dense myofibroblast populations are often found surrounding kidney cysts, their role in cyst enlargement or fibrosis in ADPKD is unclear. To clarify this, we examined the effect of myofibroblast depletion in the Pkd1RC/RC (RC/RC) mouse model of ADPKD. RC/RC;αSMAtk mice that use the ganciclovir-thymidine kinase system to selectively deplete α-smooth muscle actin expressing myofibroblasts were generated. Ganciclovir treatment for four weeks depleted myofibroblasts, reduced kidney fibrosis and preserved kidney function in these mice. Importantly, myofibroblast depletion significantly reduced cyst growth and cyst epithelial cell proliferation in RC/RC;αSMAtk mouse kidneys. Similar ganciclovir treatment did not alter cyst growth or fibrosis in wild-type or RC/RC littermates. In vitro, co-culture with myofibroblasts from the kidneys of patients with ADPKD increased 3D microcyst growth of human ADPKD cyst epithelial cells. Treatment with conditioned culture media from ADPKD kidney myofibroblasts increased microcyst growth and cell proliferation of ADPKD cyst epithelial cells. Further examination of ADPKD myofibroblast conditioned media showed high levels of protease inhibitors including PAI1, TIMP1 and 2, NGAL and TFPI-2, and treatment with recombinant PAI1 and TIMP1 increased ADPKD cyst epithelial cell proliferation in vitro. Thus, our findings show that myofibroblasts directly promote cyst epithelial cell proliferation, cyst growth and fibrosis in ADPKD kidneys, and their targeting could be a novel therapeutic strategy to treat PKD.
Collapse
Affiliation(s)
- Nidhi Dwivedi
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Abeda Jamadar
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sijo Mathew
- Department of Pharmaceutical Sciences, School of Pharmacy, North Dakota State University, Fargo, North Dakota, USA
| | - Timothy A Fields
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA; Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Reena Rao
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA; Department of Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA.
| |
Collapse
|
7
|
Chen DQ, Chen L, Guo Y, Wu XQ, Zhao TT, Zhao HL, Zhang HJ, Yan MH, Zhang GQ, Li P. Poricoic acid A suppresses renal fibroblast activation and interstitial fibrosis in UUO rats via upregulating Sirt3 and promoting β-catenin K49 deacetylation. Acta Pharmacol Sin 2022; 44:1038-1050. [PMID: 36470978 PMCID: PMC10104829 DOI: 10.1038/s41401-022-01026-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/06/2022] [Indexed: 12/12/2022] Open
Abstract
AbstractRenal interstitial fibrosis is the common pathological process of various chronic kidney diseases to end-stage renal disease. Inhibition of fibroblast activation attenuates renal interstitial fibrosis. Our previous studies show that poricoic acid A (PAA) isolated from Poria cocos is a potent anti-fibrotic agent. In the present study we investigated the effects of PAA on renal fibroblast activation and interstitial fibrosis and the underlying mechanisms. Renal interstitial fibrosis was induced in rats or mice by unilateral ureteral obstruction (UUO). UUO rats were administered PAA (10 mg·kg−1·d−1, i.g.) for 1 or 2 weeks. An in vitro model of renal fibrosis was established in normal renal kidney fibroblasts (NRK-49F cells) treated with TGF-β1. We showed that PAA treatment rescued Sirt3 expression, and significantly attenuated renal fibroblast activation and interstitial fibrosis in both the in vivo and in vitro models. In TGF-β1-treated NRK-49F cells, we demonstrated that Sirt3 deacetylated β-catenin (a key transcription factor of fibroblast activation) and then accelerated its ubiquitin-dependent degradation, thus suppressing the protein expression and promoter activity of pro-fibrotic downstream target genes (twist, snail1, MMP-7 and PAI-1) to alleviate fibroblast activation; the lysine-49 (K49) of β-catenin was responsible for Sirt3-mediated β-catenin deacetylation. In molecular docking analysis, we found the potential interaction of Sirt3 and PAA. In both in vivo and in vitro models, pharmacological activation of Sirt3 by PAA significantly suppressed renal fibroblast activation via facilitating β-catenin K49 deacetylation. In UUO mice and NRK-49F cells, Sirt3 overexpression enhanced the anti-fibrotic effect of PAA, whereas Sirt3 knockdown weakened the effect. Taken together, PAA attenuates renal fibroblast activation and interstitial fibrosis by upregulating Sirt3 and inducing β-catenin K49 deacetylation, highlighting Sirt3 functions as a promising therapeutic target of renal fibroblast activation and interstitial fibrosis.
Collapse
|
8
|
Luther JM, Fogo AB. The role of mineralocorticoid receptor activation in kidney inflammation and fibrosis. Kidney Int Suppl (2011) 2022; 12:63-68. [DOI: 10.1016/j.kisu.2021.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/15/2021] [Accepted: 11/08/2021] [Indexed: 12/20/2022] Open
|
9
|
Kintscher U, Bakris GL, Kolkhof P. Novel Non-Steroidal Mineralocorticoid Receptor Antagonists in Cardiorenal Disease. Br J Pharmacol 2021; 179:3220-3234. [PMID: 34811750 DOI: 10.1111/bph.15747] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/26/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022] Open
Abstract
Mineralocorticoid receptor (MR) antagonists (MRAs) are key agents in guideline-oriented drug therapy for cardiovascular (CV) diseases such as chronic heart failure with reduced ejection fraction (HFrEF) and resistant hypertension. Currently available steroidal MRAs are efficacious in reducing morbidity and mortality, however, they can be associated with intolerable side effects including hyperkalemia in everyday clinical practice. Recently, a new class of non-steroidal MRAs including esaxerenone, AZD9977, apararenone, KBP-5074, and finerenone have been developed with an improved benefit-risk profile and a novel indication for finerenone for diabetic kidney disease. To better understand the non-steroidal MRAs, this review provides information on the molecular pharmacology as well as relevant current preclinical and clinical data on cardiorenal outcomes. A comparative review of all compounds in the class is discussed with regard to clinical efficacy and safety as well as a perspective outlining their future use in clinical practice.
Collapse
Affiliation(s)
- Ulrich Kintscher
- Charite - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Pharmacology, Cardiovascular-Metabolic-Renal Research Center, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - George L Bakris
- Department of Medicine, American Heart Association Comprehensive Hypertension Center, University of Chicago Medicine, Chicago, IL, USA
| | - Peter Kolkhof
- Research & Early Development, Cardiovascular Research, Bayer AG, Wuppertal, Germany
| |
Collapse
|
10
|
Zhang G, Yu F, Dong R, Yu J, Luo M, Zha Y. Verbascoside alleviates renal fibrosis in unilateral ureteral obstruction rats by inhibiting macrophage infiltration. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:752-759. [PMID: 34630952 PMCID: PMC8487594 DOI: 10.22038/ijbms.2021.52759.11903] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 04/19/2021] [Indexed: 12/29/2022]
Abstract
Objective(s): To explore the effect of verbascoside on renal fibrosis in unilateral ureteral obstruction (UUO) rats. Materials and Methods: Twenty Sprague-Dawley rats were randomly distributed into sham-operated, UUO, and UUO+Verbascoside groups. After two weeks of rat model construction, urine and blood samples were collected for biochemical analysis while kidney tissues were harvested for hematoxylin and eosin (H&E), Masson’s Trichrome, and immunohistochemistry staining. Pearson coefficient was used to analyze the correlation between the two proteins. Results: Verbascoside improved UUO-induced renal dysfunction as detected by decreased serum creatinine, urea nitrogen, and urinary protein excretion rate. In UUO rats, H&E staining result revealed increased total nucleated cell number, and Masson’s Trichrome staining results showed tubular interstitial fibrosis with the deposition of collagen fibrils. Besides, expressions of fibrosis-related proteins including collagen type I (COL-I), α-smooth muscle actin (a-SMA), and tissue inhibitor of metalloproteinase 2 (TIMP2) expressed higher in the UUO group. Moreover, macrophage infiltration-related factors such as CD68+, F4/80+ cells, and suppressor of cytokine signaling-3 (SOCS3) were significantly higher in the UUO group than in sham-operated rats. However, after administration with verbascoside, the accumulation of collagen fibrils and total nucleated cell numbers were mitigated. Likewise, macrophage infiltration was extenuated and fibrosis-related proteins were down-regulated in the UUO+Verbascoside rats. Correlation analysis indicated that macrophage infiltration-related markers were related to fibrosis-related factors. Conclusion: Verbascoside could alleviate renal fibrosis in UUO rats probably through ameliorating macrophage infiltration.
Collapse
Affiliation(s)
- Guihua Zhang
- Guizhou University School of Medicine, Guizhou University, Gui Yang, Gui Zhou, China
| | - Fuxun Yu
- The NHC Key Laboratory of Pulmonary Immunological Disease, Guizhou Provincial People's Hospital, Gui Yang, Gui Zhou, China
| | - Rong Dong
- Guizhou University School of Medicine, Guizhou University, Gui Yang, Gui Zhou, China.,Department of Nephrology, Guizhou Provincial People's Hospital, Gui Yang, Gui Zhou, China
| | - Jiali Yu
- Guizhou University School of Medicine, Guizhou University, Gui Yang, Gui Zhou, China.,Department of Nephrology, Guizhou Provincial People's Hospital, Gui Yang, Gui Zhou, China
| | - Meng Luo
- Guizhou University School of Medicine, Guizhou University, Gui Yang, Gui Zhou, China.,Department of Thoracic Surgery, Guizhou Provincial People's Hospital, Gui Yang, Gui Zhou, China
| | - Yan Zha
- Guizhou University School of Medicine, Guizhou University, Gui Yang, Gui Zhou, China.,Department of Nephrology, Guizhou Provincial People's Hospital, Gui Yang, Gui Zhou, China
| |
Collapse
|
11
|
Yan H, Xu J, Xu Z, Yang B, Luo P, He Q. Defining therapeutic targets for renal fibrosis: Exploiting the biology of pathogenesis. Biomed Pharmacother 2021; 143:112115. [PMID: 34488081 DOI: 10.1016/j.biopha.2021.112115] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 08/11/2021] [Accepted: 08/19/2021] [Indexed: 01/14/2023] Open
Abstract
Renal fibrosis is a failed wound-healing process of the kidney tissue after chronic, sustained injury, which is a common pathway and pathological marker of virtually every type of chronic kidney disease (CKD), regardless of cause. However, there is a lack of effective treatment specifically targeting against renal fibrosis per se to date. The main pathological feature of renal fibrosis is the massive activation and proliferation of renal fibroblasts and the excessive synthesis and secretion of extracellular matrix (ECM) deposited in the renal interstitium, leading to structural damage, impairment of renal function, and eventually end-stage renal disease. In this review, we summarize recent advancements regarding the participation and interaction of many types of kidney residents and infiltrated cells during renal fibrosis, attempt to comprehensively discuss the mechanism of renal fibrosis from the cellular level and conclude by highlighting novel therapeutic targets and approaches for development of new treatments for patients with renal fibrosis.
Collapse
Affiliation(s)
- Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiangxin Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
12
|
Li SS, Sun Q, Hua MR, Suo P, Chen JR, Yu XY, Zhao YY. Targeting the Wnt/β-Catenin Signaling Pathway as a Potential Therapeutic Strategy in Renal Tubulointerstitial Fibrosis. Front Pharmacol 2021; 12:719880. [PMID: 34483931 PMCID: PMC8415231 DOI: 10.3389/fphar.2021.719880] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022] Open
Abstract
The Wnt/β-catenin signaling pathway plays important roles in embryonic development and tissue homeostasis. Wnt signaling is induced, and β-catenin is activated, associated with the development and progression of renal fibrosis. Wnt/β-catenin controls the expression of various downstream mediators such as snail1, twist, matrix metalloproteinase-7, plasminogen activator inhibitor-1, transient receptor potential canonical 6, and renin-angiotensin system components in epithelial cells, fibroblast, and macrophages. In addition, Wnt/β-catenin is usually intertwined with other signaling pathways to promote renal interstitial fibrosis. Actually, given the crucial of Wnt/β-catenin signaling in renal fibrogenesis, blocking this signaling may benefit renal interstitial fibrosis. There are several antagonists of Wnt signaling that negatively control Wnt activation, and these include soluble Fzd-related proteins, the family of Dickkopf 1 proteins, Klotho and Wnt inhibitory factor-1. Furthermore, numerous emerging small-molecule β-catenin inhibitors cannot be ignored to prevent and treat renal fibrosis. Moreover, we reviewed the knowledge focusing on anti-fibrotic effects of natural products commonly used in kidney disease by inhibiting the Wnt/β-catenin signaling pathway. Therefore, in this review, we summarize recent advances in the regulation, downstream targets, role, and mechanisms of Wnt/β-catenin signaling in renal fibrosis pathogenesis. We also discuss the therapeutic potential of targeting this pathway to treat renal fibrosis; this may shed new insights into effective treatment strategies to prevent and treat renal fibrosis.
Collapse
Affiliation(s)
- Shan-Shan Li
- Department of Nephrology, Shaanxi Traditional Chinese Medicine Hospital, Xi'an, China.,The First School of Clinical Medicine, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| | - Qian Sun
- Department of Nephrology, Shaanxi Traditional Chinese Medicine Hospital, Xi'an, China.,The First School of Clinical Medicine, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| | - Meng-Ru Hua
- Faculty of Life Science and Medicine, Northwest University, Xi'an, China
| | - Ping Suo
- Faculty of Life Science and Medicine, Northwest University, Xi'an, China
| | - Jia-Rong Chen
- Department of Clinical Pharmacy, Affiliated Hospital of Chengdu University, Chengdu, China
| | - Xiao-Yong Yu
- Department of Nephrology, Shaanxi Traditional Chinese Medicine Hospital, Xi'an, China
| | - Ying-Yong Zhao
- Faculty of Life Science and Medicine, Northwest University, Xi'an, China
| |
Collapse
|
13
|
Gifford CC, Lian F, Tang J, Costello A, Goldschmeding R, Samarakoon R, Higgins PJ. PAI-1 induction during kidney injury promotes fibrotic epithelial dysfunction via deregulation of klotho, p53, and TGF-β1-receptor signaling. FASEB J 2021; 35:e21725. [PMID: 34110636 DOI: 10.1096/fj.202002652rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/05/2021] [Accepted: 05/24/2021] [Indexed: 12/13/2022]
Abstract
Renal fibrosis leads to chronic kidney disease, which affects over 15% of the U.S. population. PAI-1 is highly upregulated in the tubulointerstitial compartment in several common nephropathies and PAI-1 global ablation affords protection from fibrogenesis in mice. The precise contribution of renal tubular PAI-1 induction to disease progression, however, is unknown and surprisingly, appears to be independent of uPA inhibition. Human renal epithelial (HK-2) cells engineered to stably overexpress PAI-1 underwent dedifferentiation (E-cadherin loss, gain of vimentin), G2/M growth arrest (increased p-Histone3, p21), and robust induction of fibronectin, collagen-1, and CCN2. These cells are also susceptible to apoptosis (elevated cleaved caspase-3, annexin-V positivity) compared to vector controls, demonstrating a previously unknown role for PAI-1 in tubular dysfunction. Persistent PAI-1 expression results in a loss of klotho expression, p53 upregulation, and increases in TGF-βRI/II levels and SMAD3 phosphorylation. Ectopic restoration of klotho in PAI-1-transductants attenuated fibrogenesis and reversed the proliferative defects, implicating PAI-1 in klotho loss in renal disease. Genetic suppression of p53 reversed the PA1-1-driven maladaptive repair, moreover, confirming a pathogenic role for p53 upregulation in this context and uncovering a novel role for PAI-1 in promoting renal p53 signaling. TGF-βRI inhibition also attenuated PAI-1-initiated epithelial dysfunction, independent of TGF-β1 ligand synthesis. Thus, PAI-1 promotes tubular dysfunction via klotho reduction, p53 upregulation, and activation of the TGF-βRI-SMAD3 axis. Since klotho is an upstream regulator of both PAI-1-mediated p53 induction and SMAD3 signaling, targeting tubular PAI-1 expression may provide a novel, multi-level approach to the therapy of CKD.
Collapse
Affiliation(s)
- Cody C Gifford
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, USA
| | - Fei Lian
- Division of Urology, Albany Medical College, Albany, NY, USA
| | - Jiaqi Tang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, USA
| | - Angelica Costello
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, USA
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Rohan Samarakoon
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, USA
| | - Paul J Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, Albany, NY, USA.,Division of Urology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
14
|
Zhong J, Yang HC, Yermalitsky V, Shelton EL, Otsuka T, Wiese CB, May-Zhang LS, Banan B, Abumrad N, Huang J, Cavnar AB, Kirabo A, Yancey PG, Fogo AB, Vickers KC, Linton MF, Davies SS, Kon V. Kidney injury-mediated disruption of intestinal lymphatics involves dicarbonyl-modified lipoproteins. Kidney Int 2021; 100:585-596. [PMID: 34102217 DOI: 10.1016/j.kint.2021.05.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 04/06/2021] [Accepted: 05/06/2021] [Indexed: 12/24/2022]
Abstract
Kidney disease affects intestinal structure and function. Although intestinal lymphatics are central in absorption and remodeling of dietary and synthesized lipids/lipoproteins, little is known about how kidney injury impacts the intestinal lymphatic network, or lipoproteins transported therein. To study this, we used puromycin aminoglycoside-treated rats and NEP25 transgenic mice to show that proteinuric injury expanded the intestinal lymphatic network, activated lymphatic endothelial cells and increased mesenteric lymph flow. The lymph was found to contain increased levels of cytokines, immune cells, and isolevuglandin (a highly reactive dicarbonyl) and to have a greater output of apolipoprotein AI. Plasma levels of cytokines and isolevuglandin were not changed. However, isolevuglandin was also increased in the ileum of proteinuric animals, and intestinal epithelial cells exposed to myeloperoxidase produced more isolevuglandin. Apolipoprotein AI modified by isolevuglandin directly increased lymphatic vessel contractions, activated lymphatic endothelial cells, and enhanced the secretion of the lymphangiogenic promoter vascular endothelial growth factor-C by macrophages. Inhibition of isolevuglandin synthesis by a carbonyl scavenger reduced intestinal isolevuglandin adduct level and lymphangiogenesis. Thus, our data reveal a novel mediator, isolevuglandin modified apolipoprotein AI, and uncover intestinal lymphatic network structure and activity as a new pathway in the crosstalk between kidney and intestine that may contribute to the adverse impact of kidney disease on other organs.
Collapse
Affiliation(s)
- Jianyong Zhong
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Hai-Chun Yang
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| | - Valery Yermalitsky
- Department of Clinical Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | - Elaine L Shelton
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tadashi Otsuka
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Carrie B Wiese
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Linda S May-Zhang
- Department of Clinical Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | - Babak Banan
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Naji Abumrad
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jiansheng Huang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ashley B Cavnar
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Annet Kirabo
- Department of Clinical Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | - Patricia G Yancey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Agnes B Fogo
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kasey C Vickers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - MacRae F Linton
- Department of Clinical Pharmacology, Vanderbilt University, Nashville, Tennessee, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Sean S Davies
- Department of Clinical Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | - Valentina Kon
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee, USA.
| |
Collapse
|
15
|
Jaffar J, Glaspole I, Symons K, Westall G. Inhibition of NF-κB by ACT001 reduces fibroblast activity in idiopathic pulmonary fibrosis. Biomed Pharmacother 2021; 138:111471. [PMID: 33730605 DOI: 10.1016/j.biopha.2021.111471] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 02/18/2021] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease of unknown etiology and poor prognosis. In IPF, aberrant extracellular matrix production by activated, hyperproliferative fibroblasts drives disease progression but the exact mechanisms by which this occurs remains undefined. The transcription factor nuclear factor kappa-B (NF-ĸB) has been suggested as a potential therapeutic target in IPF and therefore the aim of this study was to investigate the efficacy of ACT001, an NF-ĸB inhibitor, on primary fibroblasts derived from patients with and without IPF. Primary lung fibroblasts derived from eight patients with IPF and eight age-matched non-diseased controls (NDC) were treated with 0-10 µM ACT001 and the effects on fibroblast activity (viability and proliferation, fibroblast-to-myofibroblast transition, fibronectin expression), interleukin (IL)-6 and IL-8 cytokine release were quantified. ACT001 inhibited fibroblast activity in a concentration-dependent manner in both groups of fibroblasts. ACT001 inhibited IL-6 but not IL-8 production in unstimulated fibroblasts. ACT001 is a water-soluble compound with a stable half-life in plasma, thus making it an attractive candidate for further investigation as a therapeutic in IPF. This study adds to the growing body of literature that demonstrates anti-fibrotic activity of NF-ĸB inhibition in the context of IPF.
Collapse
Affiliation(s)
- Jade Jaffar
- Department of Respiratory Medicine, The Alfred Hospital, 99 Commercial Rd, Melbourne, VIC 3000, Australia; Department of Immunology and Pathology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia; N.M.H.R.C. Centre of Research Excellence in Pulmonary Fibrosis, Australia.
| | - Ian Glaspole
- Department of Respiratory Medicine, The Alfred Hospital, 99 Commercial Rd, Melbourne, VIC 3000, Australia; Department of Immunology and Pathology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia; N.M.H.R.C. Centre of Research Excellence in Pulmonary Fibrosis, Australia
| | - Karen Symons
- Department of Respiratory Medicine, The Alfred Hospital, 99 Commercial Rd, Melbourne, VIC 3000, Australia
| | - Glen Westall
- Department of Respiratory Medicine, The Alfred Hospital, 99 Commercial Rd, Melbourne, VIC 3000, Australia; Department of Immunology and Pathology, Monash University, 89 Commercial Rd, Melbourne, VIC 3004, Australia; N.M.H.R.C. Centre of Research Excellence in Pulmonary Fibrosis, Australia
| |
Collapse
|
16
|
Rattanavirotkul N, Kirschner K, Chandra T. Induction and transmission of oncogene-induced senescence. Cell Mol Life Sci 2021; 78:843-852. [PMID: 32936311 PMCID: PMC7897614 DOI: 10.1007/s00018-020-03638-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 07/27/2020] [Accepted: 09/03/2020] [Indexed: 12/19/2022]
Abstract
Senescence is a cellular stress response triggered by diverse stressors, including oncogene activation, where it serves as a bona-fide tumour suppressor mechanism. Senescence can be transmitted to neighbouring cells, known as paracrine secondary senescence. Secondary senescence was initially described as a paracrine mechanism, but recent evidence suggests a more complex scenario involving juxtacrine communication between cells. In addition, single-cell studies described differences between primary and secondary senescent end-points, which have thus far not been considered functionally distinct. Here we discuss emerging concepts in senescence transmission and heterogeneity in primary and secondary senescence on a cellular and organ level.
Collapse
Affiliation(s)
- Nattaphong Rattanavirotkul
- Chakri Naruebodindra Medical Institute, Ramathibodi Medical School, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 111, Bang Pla, Bang Phli, Samut Prakan, 10540, Thailand.
| | - Kristina Kirschner
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1BD, UK.
| | - Tamir Chandra
- MRC Human Genetics Unit, MRC Institute of Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
17
|
WNT-β-catenin signalling - a versatile player in kidney injury and repair. Nat Rev Nephrol 2020; 17:172-184. [PMID: 32989282 DOI: 10.1038/s41581-020-00343-w] [Citation(s) in RCA: 221] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2020] [Indexed: 12/11/2022]
Abstract
The WNT-β-catenin system is an evolutionary conserved signalling pathway that is of particular importance for morphogenesis and cell organization during embryogenesis. The system is usually suppressed in adulthood; however, it can be re-activated in organ injury and regeneration. WNT-deficient mice display severe kidney defects at birth. Transient WNT-β-catenin activation stimulates tissue regeneration after acute kidney injury, whereas sustained (uncontrolled) WNT-β-catenin signalling promotes kidney fibrosis in chronic kidney disease (CKD), podocyte injury and proteinuria, persistent tissue damage during acute kidney injury and cystic kidney diseases. Additionally, WNT-β-catenin signalling is involved in CKD-associated vascular calcification and mineral bone disease. The WNT-β-catenin pathway is tightly regulated, for example, by proteins of the Dickkopf (DKK) family. In particular, DKK3 is released by 'stressed' tubular epithelial cells; DKK3 drives kidney fibrosis and is associated with short-term risk of CKD progression and acute kidney injury. Thus, targeting the WNT-β-catenin pathway might represent a promising therapeutic strategy in kidney injury and associated complications.
Collapse
|
18
|
Neural transcription factor Pou4f1 promotes renal fibrosis via macrophage-myofibroblast transition. Proc Natl Acad Sci U S A 2020; 117:20741-20752. [PMID: 32788346 DOI: 10.1073/pnas.1917663117] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Unresolved inflammation can lead to tissue fibrosis and impaired organ function. Macrophage-myofibroblast transition (MMT) is one newly identified mechanism by which ongoing chronic inflammation causes progressive fibrosis in different forms of kidney disease. However, the mechanisms underlying MMT are still largely unknown. Here, we discovered a brain-specific homeobox/POU domain protein Pou4f1 (Brn3a) as a specific regulator of MMT. Interestingly, we found that Pou4f1 is highly expressed by macrophages undergoing MMT in sites of fibrosis in human and experimental kidney disease, identified by coexpression of the myofibroblast marker, α-SMA. Unexpectedly, Pou4f1 expression peaked in the early stage in renal fibrogenesis in vivo and during MMT of bone marrow-derived macrophages (BMDMs) in vitro. Mechanistically, chromatin immunoprecipitation (ChIP) assay identified that Pou4f1 is a Smad3 target and the key downstream regulator of MMT, while microarray analysis defined a Pou4f1-dependent fibrogenic gene network for promoting TGF-β1/Smad3-driven MMT in BMDMs at the transcriptional level. More importantly, using two mouse models of progressive renal interstitial fibrosis featuring the MMT process, we demonstrated that adoptive transfer of TGF-β1-stimulated BMDMs restored both MMT and renal fibrosis in macrophage-depleted mice, which was prevented by silencing Pou4f1 in transferred BMDMs. These findings establish a role for Pou4f1 in MMT and renal fibrosis and suggest that Pou4f1 may be a therapeutic target for chronic kidney disease with progressive renal fibrosis.
Collapse
|
19
|
Abstract
Chronic kidney disease (CKD) is a devastating condition that is reaching epidemic levels owing to the increasing prevalence of diabetes mellitus, hypertension and obesity, as well as ageing of the population. Regardless of the underlying aetiology, CKD is slowly progressive and leads to irreversible nephron loss, end-stage renal disease and/or premature death. Factors that contribute to CKD progression include parenchymal cell loss, chronic inflammation, fibrosis and reduced regenerative capacity of the kidney. Current therapies have limited effectiveness and only delay disease progression, underscoring the need to develop novel therapeutic approaches to either stop or reverse progression. Preclinical studies have identified several approaches that reduce fibrosis in experimental models, including targeting cytokines, transcription factors, developmental and signalling pathways and epigenetic modulators, particularly microRNAs. Some of these nephroprotective strategies are now being tested in clinical trials. Lessons learned from the failure of clinical studies of transforming growth factor β1 (TGFβ1) blockade underscore the need for alternative approaches to CKD therapy, as strategies that target a single pathogenic process may result in unexpected negative effects on simultaneously occurring processes. Additional promising avenues include preventing tubular cell injury and anti-fibrotic therapies that target activated myofibroblasts, the main collagen-producing cells.
Collapse
|
20
|
Knoppert SN, Valentijn FA, Nguyen TQ, Goldschmeding R, Falke LL. Cellular Senescence and the Kidney: Potential Therapeutic Targets and Tools. Front Pharmacol 2019; 10:770. [PMID: 31354486 PMCID: PMC6639430 DOI: 10.3389/fphar.2019.00770] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/14/2019] [Indexed: 01/10/2023] Open
Abstract
Chronic kidney disease (CKD) is an increasing health burden (affecting approximately 13.4% of the population). Currently, no curative treatment options are available and treatment is focused on limiting the disease progression. The accumulation of senescent cells has been implicated in the development of kidney fibrosis by limiting tissue rejuvenation and through the secretion of pro-fibrotic and pro-inflammatory mediators termed as the senescence-associated secretory phenotype. The clearance of senescent cells in aging models results in improved kidney function, which shows promise for the options of targeting senescent cells in CKD. There are several approaches for the development of “senotherapies”, the most rigorous of which is the elimination of senescent cells by the so-called senolytic drugs either newly developed or repurposed for off-target effects in terms of selectively inducing apoptosis in senescent cells. Several chemotherapeutics and checkpoint inhibitors currently used in daily oncological practice show senolytic properties. However, the applicability of such senolytic compounds for the treatment of renal diseases has hardly been investigated. A serious concern is that systemic side effects will limit the use of senolytics for kidney fibrosis. Specifically targeting senescent cells and/or targeted drug delivery to the kidney might circumvent these side effects. In this review, we discuss the connection between CKD and senescence, the pharmacological options for targeting senescent cells, and the means to specifically target the kidney.
Collapse
Affiliation(s)
- Sebastian N Knoppert
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Floris A Valentijn
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Tri Q Nguyen
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Lucas L Falke
- Department of Pathology, University Medical Center Utrecht, Utrecht, Netherlands.,Department of Internal Medicine, Diakonessenhuis, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|