151
|
Uremic Sarcopenia: Clinical Evidence and Basic Experimental Approach. Nutrients 2020; 12:nu12061814. [PMID: 32570738 PMCID: PMC7353433 DOI: 10.3390/nu12061814] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/27/2020] [Accepted: 06/16/2020] [Indexed: 12/17/2022] Open
Abstract
Sustained physical activity extends healthy life years while a lower activity due to sarcopenia can reduce them. Sarcopenia is defined as a decrease in skeletal muscle mass and strength due not only to aging, but also from a variety of debilitating chronic illnesses such as cancer and heart failure. Patients with chronic kidney disease (CKD), who tend to be cachexic and in frail health, may develop uremic sarcopenia or uremic myopathy due to an imbalance between muscle protein synthesis and catabolism. Here, we review clinical evidence indicating reduced physical activity as renal function deteriorates and explore evidence-supported therapeutic options focusing on nutrition and physical training. In addition, although sarcopenia is a clinical concept and difficult to recapitulate in basic research, several in vivo approaches have been attempted, such as rodent subtotal nephrectomy representing both renal dysfunction and muscle weakness. This review highlights molecular mechanisms and promising interventions for uremic sarcopenia that were revealed through basic research. Extensive study is still needed to cast light on the many aspects of locomotive organ impairments in CKD and explore the ways that diet and exercise therapies can improve both outcomes and quality of life at every level.
Collapse
|
152
|
Shiva N, Sharma N, Kulkarni YA, Mulay SR, Gaikwad AB. Renal ischemia/reperfusion injury: An insight on in vitro and in vivo models. Life Sci 2020; 256:117860. [PMID: 32534037 DOI: 10.1016/j.lfs.2020.117860] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/21/2020] [Accepted: 05/26/2020] [Indexed: 02/08/2023]
Abstract
Optimal tissue oxygenation is essential for its normal function. Suboptimal oxygenation or ischemia contributes to increased mortalities during various pathological conditions such as stroke, acute kidney injury (AKI), cardiac failure. Despite the rapid progression of renal tissue injury, the mechanism underlying renal ischemia/reperfusion injury (IRI) remains highly unclear. Experimental in vitro and in vivo models epitomizing the fundamental process is critical to the research of the pathogenesis of IRI and the development of plausible therapeutics. In this review, we describe the in vitro and in vivo models of IRI, ranges from proximal tubular cell lines to surgery-based animal models like clamping of both renal pedicles (bilateral IRI), clamping of one renal pedicle (unilateral IRI), clamping of one/or both renal arteries/or vein, or unilateral IRI with contralateral nephrectomy (uIRIx). Also, advanced technologies like three-dimensional kidney organoids, kidney-on-a-chip are explained. This review provides thoughtful information for establishing reliable and pertinent models for studying IRI-associated acute renal pathologies.
Collapse
Affiliation(s)
- Niharika Shiva
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Nisha Sharma
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| | - Shrikant R Mulay
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|
153
|
Zhu H, Liao J, Zhou X, Hong X, Song D, Hou FF, Liu Y, Fu H. Tenascin-C promotes acute kidney injury to chronic kidney disease progression by impairing tubular integrity via αvβ6 integrin signaling. Kidney Int 2020; 97:1017-1031. [PMID: 32245660 PMCID: PMC8112450 DOI: 10.1016/j.kint.2020.01.026] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 12/20/2019] [Accepted: 01/09/2020] [Indexed: 12/28/2022]
Abstract
Tenascin-C is an extracellular matrix glycoprotein that plays a critical role in kidney fibrosis by orchestrating a fibrogenic niche. Here, we demonstrate that tenascin-C is a biomarker and a mediator of kidney fibrogenesis by impairing tubular integrity. Tenascin-C was found to be increased in kidney biopsies from patients with chronic kidney disease (CKD). In a cohort of 225 patients with CKD, the urinary tenascin-C level was markedly elevated, compared to 39 healthy individuals. Moreover, the level of urinary tenascin-C in CKD was correlated with the severity of kidney dysfunction and fibrosis. In mouse model of acute kidney injury-to-CKD induced by ischemia/reperfusion, depletion of tenascin-C preserved tubular integrity and ameliorated renal fibrotic lesions. In vitro, tenascin-C impaired tubular cell integrity by inducing partial epithelial-mesenchymal transition. Using decellularized kidney tissue scaffolds, we found that tenascin-C-enriched scaffolds facilitated tubular epithelial-mesenchymal transition ex vivo. Mechanistically, tenascin-C specifically induced integrins αvβ6 in tubular cells and activated focal adhesion kinase (FAK). Blocking αvβ6 integrins or inhibition of FAK restored tubular integrity by repressing epithelial-mesenchymal transition and alleviated kidney fibrosis. Thus, our studies underscore that tenascin-C is a noninvasive biomarker of kidney fibrogenesis and a pathogenic mediator that impairs tubular integrity. Hence, blockade of the tenascin-C/αvβ6 integrin/FAK signal cascade may be a novel strategy for therapeutic intervention of kidney fibrosis.
Collapse
Affiliation(s)
- Haili Zhu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinlin Liao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xianke Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xue Hong
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dongyan Song
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China; Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| | - Haiyan Fu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
154
|
Ishiuchi N, Nakashima A, Doi S, Yoshida K, Maeda S, Kanai R, Yamada Y, Ike T, Doi T, Kato Y, Masaki T. Hypoxia-preconditioned mesenchymal stem cells prevent renal fibrosis and inflammation in ischemia-reperfusion rats. Stem Cell Res Ther 2020; 11:130. [PMID: 32197638 PMCID: PMC7083035 DOI: 10.1186/s13287-020-01642-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 02/12/2020] [Accepted: 03/10/2020] [Indexed: 12/17/2022] Open
Abstract
Background Mesenchymal stem cells (MSCs) have been reported to promote the regeneration of injured tissue via their paracrine abilities, which are enhanced by hypoxic preconditioning. In this study, we examined the therapeutic efficacy of hypoxia-preconditioned MSCs on renal fibrosis and inflammation in rats with ischemia-reperfusion injury (IRI). Methods MSCs derived from rats and humans were incubated in 1% O2 conditions (1%O2 MSCs) for 24 h. After IRI, 1%O2 MSCs or MSCs cultured under normoxic conditions (21%O2 MSCs) were injected through the abdominal aorta. At 7 or 21 days post-injection, the rats were sacrificed and their kidneys were analyzed. In in vitro experiments, we examined whether 1%O2 MSCs enhanced the ability to produce anti-fibrotic humoral factors using transforming growth factor (TGF)-β1-stimulated HK-2 cells incubated with conditioned medium from MSCs. Results Administration of rat 1%O2 MSCs (1%O2 rMSCs) attenuated renal fibrosis and inflammation more significantly than rat 21%O2 MSCs. Notably, human 1%O2 MSCs (1%O2 hMSCs) also attenuated renal fibrosis to the same extent as 1%O2 rMSCs. Flow cytometry showed that 1%O2 hMSCs did not change human leukocyte antigen expression. Further in vitro experiments revealed that conditioned medium from 1%O2 MSCs further suppressed TGF-β1-induced fibrotic changes in HK-2 cells compared with 21%O2 MSCs. Hypoxic preconditioning enhanced vascular endothelial growth factor (VEGF) and hepatocyte growth factor (HGF) secretion. Interestingly, VEGF knockdown in 1%O2 MSCs attenuated HGF secretion and the inhibition of TGF-β1-induced fibrotic changes in HK-2 cells. In addition, VEGF knockdown in 1%O2 hMSCs reduced the anti-fibrotic effect in IRI rats. Conclusions Our results indicate that hypoxia-preconditioned MSCs are useful as an allogeneic transplantation cell therapy to prevent renal fibrosis and inflammation.
Collapse
Affiliation(s)
- Naoki Ishiuchi
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Ayumu Nakashima
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan. .,Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.
| | - Shigehiro Doi
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Ken Yoshida
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Satoshi Maeda
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.,TWOCELLS Company, Limited, 16-35 Hijiyama-honmachi, Minami-ku, Hiroshima, 732-0816, Japan
| | - Ryo Kanai
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yumi Yamada
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Takeshi Ike
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Toshiki Doi
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yukio Kato
- Department of Stem Cell Biology and Medicine, Graduate School of Biomedical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.,TWOCELLS Company, Limited, 16-35 Hijiyama-honmachi, Minami-ku, Hiroshima, 732-0816, Japan
| | - Takao Masaki
- Department of Nephrology, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| |
Collapse
|
155
|
Polichnowski AJ, Griffin KA, Licea-Vargas H, Lan R, Picken MM, Long J, Williamson GA, Rosenberger C, Mathia S, Venkatachalam MA, Bidani AK. Pathophysiology of unilateral ischemia-reperfusion injury: importance of renal counterbalance and implications for the AKI-CKD transition. Am J Physiol Renal Physiol 2020; 318:F1086-F1099. [PMID: 32174143 DOI: 10.1152/ajprenal.00590.2019] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Unilateral ischemia-reperfusion (UIR) injury leads to progressive renal atrophy and tubulointerstitial fibrosis (TIF) and is commonly used to investigate the pathogenesis of the acute kidney injury-chronic kidney disease transition. Although it is well known that contralateral nephrectomy (CNX), even 2 wk post-UIR injury, can improve recovery, the physiological mechanisms and tubular signaling pathways mediating such improved recovery remain poorly defined. Here, we examined the renal hemodynamic and tubular signaling pathways associated with UIR injury and its reversal by CNX. Male Sprague-Dawley rats underwent left UIR or sham UIR and 2 wk later CNX or sham CNX. Blood pressure, left renal blood flow (RBF), and total glomerular filtration rate were assessed in conscious rats for 3 days before and over 2 wk after CNX or sham CNX. In the presence of a contralateral uninjured kidney, left RBF was lower (P < 0.05) from 2 to 4 wk following UIR (3.6 ± 0.3 mL/min) versus sham UIR (9.6 ± 0.3 mL/min). Without CNX, extensive renal atrophy, TIF, and tubule dedifferentiation, but minimal pimonidazole and hypoxia-inducible factor-1α positivity in tubules, were present at 4 wk post-UIR injury. Conversely, CNX led (P < 0.05) to sustained increases in left RBF (6.2 ± 0.6 mL/min) that preceded the increases in glomerular filtration rate. The CNX-induced improvement in renal function was associated with renal hypertrophy, more redifferentiated tubules, less TIF, and robust pimonidazole and hypoxia-inducible factor-1α staining in UIR injured kidneys. Thus, contrary to expectations, indexes of hypoxia are not observed with the extensive TIF at 4 wk post-UIR injury in the absence of CNX but are rather associated with the improved recovery of renal function and structure following CNX.
Collapse
Affiliation(s)
- Aaron J Polichnowski
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee.,Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee.,Renal Section, Department of Medicine, Edward Hines Jr. Veterans Administration Hospital, Hines, Illinois.,Division of Nephrology, Department of Medicine, Loyola University Medical Center, Maywood, Illinois
| | - Karen A Griffin
- Renal Section, Department of Medicine, Edward Hines Jr. Veterans Administration Hospital, Hines, Illinois.,Division of Nephrology, Department of Medicine, Loyola University Medical Center, Maywood, Illinois
| | - Hector Licea-Vargas
- Renal Section, Department of Medicine, Edward Hines Jr. Veterans Administration Hospital, Hines, Illinois.,Division of Nephrology, Department of Medicine, Loyola University Medical Center, Maywood, Illinois
| | - Rongpei Lan
- Department of Pathology, University of Texas Health Science Center, San Antonio, Texas
| | - Maria M Picken
- Department of Pathology, Loyola University Medical Center, Maywood, Illinois
| | - Jainrui Long
- Department of Electrical and Computer Engineering, Illinois Institute of Technology, Chicago, Illinois
| | - Geoffrey A Williamson
- Department of Electrical and Computer Engineering, Illinois Institute of Technology, Chicago, Illinois
| | - Christian Rosenberger
- Department of Nephrology and Medical Intensive Care, Charité Universitaetsmedizin, Berlin, Germany
| | - Susanne Mathia
- Department of Nephrology and Medical Intensive Care, Charité Universitaetsmedizin, Berlin, Germany
| | | | - Anil K Bidani
- Renal Section, Department of Medicine, Edward Hines Jr. Veterans Administration Hospital, Hines, Illinois.,Division of Nephrology, Department of Medicine, Loyola University Medical Center, Maywood, Illinois
| |
Collapse
|
156
|
Liu L, Deng Y, Cai Y, Lu P, Guo Y, Zhang C, Li Q, Zhang T, Han M, Xu G. Ablation of Gsa impairs renal tubule proliferation after injury via CDK2/cyclin E. Am J Physiol Renal Physiol 2020; 318:F793-F803. [PMID: 32036696 DOI: 10.1152/ajprenal.00367.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Acute kidney injury has a high global morbidity associated with an increased risk of death and chronic kidney disease. Renal tubular epithelial cell regeneration following injury may be a decisive factor in renal repair or the progression of acute kidney injury to chronic kidney disease, but the underlying mechanism of abnormal renal tubular repair remains unclear. In the present study, we investigated the role of heterotrimeric G stimulatory protein α-subunit (Gsa) in renal tubular epithelial cell regeneration. We generated renal tubule epithelium-specific Gsa knockout (GsaKspKO) mice to show the essential role of Gsa in renal tubular epithelial cell regeneration in two AKI models: acute aristolochic acid nephropathy (AAN) and unilateral ischemia-reperfusion injury (UIRI). GsaKspKO mice developed more severe renal impairment after AAN and UIRI, higher serum creatinine levels, and more substantial tubular necrosis than wild-type mice. More importantly, Gsa inactivation impaired renal tubular epithelial cell proliferation by reducing bromodeoxyuridine+ cell numbers in the AAN model and inhibiting cyclin-dependent kinase 2/cyclin E1 expression in the UIRI model. This reduced proliferation was further supported in vitro with Gsa-targeting siRNA. Downregulation of Gsa inhibited tubular epithelial cell proliferation in HK-2 and mIMCD-3 cells. Furthermore, Gsa downregulation inhibited cyclin-dependent kinase 2/cyclin E1 expression, which was dependent on the Raf-MEK-ERK signaling pathway. In conclusion, Gsa is required for tubular epithelial cell regeneration during kidney repair after AKI. Loss of Gsa impairs renal tubular epithelial cell regeneration by blocking the Raf-MEK-ERK pathway.
Collapse
Affiliation(s)
- Lele Liu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanjun Deng
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Cai
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pingfan Lu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiyan Guo
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunjiang Zhang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Li
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianjing Zhang
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Han
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Xu
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
157
|
Characterization of Matricellular Protein Expression Signatures in Mechanistically Diverse Mouse Models of Kidney Injury. Sci Rep 2019; 9:16736. [PMID: 31723159 PMCID: PMC6854083 DOI: 10.1038/s41598-019-52961-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/24/2019] [Indexed: 12/14/2022] Open
Abstract
Fibrosis is the most common pathophysiological manifestation of Chronic Kidney Disease (CKD). It is defined as excessive deposition of extracellular matrix (ECM) proteins. Embedded within the ECM are a family of proteins called Matricellular Proteins (MCPs), which are typically expressed during chronic pathologies for ECM processing. As such, identifying potential MCPs in the pathological secretome of a damaged kidney could serve as diagnostic/therapeutic targets of fibrosis. Using published RNA-Seq data from two kidney injury mouse models of different etiologies, Folic Acid (FA) and Unilateral Ureteral Obstruction (UUO), we compared and contrasted the expression profile of various members from well-known MCP families during the Acute and Fibrotic injury phases. As a result, we identified common and distinct MCP expression signatures between both injury models. Bioinformatic analysis of their differentially expressed MCP genes revealed similar top annotation clusters from Molecular Function and Biological Process networks, which are those commonly involved in fibrosis. Using kidney lysates from FA- and UUO-injured mice, we selected MCP genes from our candidate list to confirm mRNA expression by Western Blot, which correlated with injury progression. Understanding the expressions of MCPs will provide important insight into the processes of kidney repair, and may validate MCPs as biomarkers and/or therapeutic targets of CKD.
Collapse
|
158
|
Zhuo H, Zhou D, Wang Y, Mo H, Yu Y, Liu Y. Sonic hedgehog selectively promotes lymphangiogenesis after kidney injury through noncanonical pathway. Am J Physiol Renal Physiol 2019; 317:F1022-F1033. [PMID: 31411078 PMCID: PMC6843033 DOI: 10.1152/ajprenal.00077.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 07/24/2019] [Accepted: 08/01/2019] [Indexed: 02/08/2023] Open
Abstract
Kidney fibrosis is associated with an increased lymphangiogenesis, characterized by the formation and expansion of new lymphatic vessels. However, the trigger and underlying mechanism responsible for the growth of lymphatic vessels in diseased kidney remain poorly defined. Here, we report that tubule-derived sonic hedgehog (Shh) ligand is a novel lymphangiogenic factor that plays a crucial role in mediating lymphatic endothelial cell proliferation and expansion. Shh was induced in renal tubular epithelium in various models of fibrotic chronic kidney disease, and this was accompanied by an expansion of lymphatic vessels in adjacent areas. In vitro, Shh selectively promoted the proliferation of human dermal lymphatic endothelial cells (HDLECs) but not human umbilical vein endothelial cells, as assessed by cell counting, MTT assay, and bromodeoxyuridine incorporation. Shh also induced the expression of vascular endothelial growth factor receptor-3, cyclin D1, and proliferating cell nuclear antigen in HDLECs. Shh did not affect the expression of Gli1, the downstream target and readout of canonical hedgehog signaling, but activated ERK-1/2 in HDLECs. Inhibition of Smoothened with small-molecule inhibitor or blockade of ERK-1/2 activation abolished the lymphatic endothelial cell proliferation induced by Shh. In vivo, inhibition of Smoothened also repressed lymphangiogenesis and attenuated renal fibrosis. This study identifies Shh as a novel mitogen that selectively promotes lymphatic, but not vascular, endothelial cell proliferation and suggests that tubule-derived Shh plays an essential role in mediating lymphangiogenesis after kidney injury.
Collapse
Affiliation(s)
- Hui Zhuo
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Dong Zhou
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yuanyuan Wang
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Hongyan Mo
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ying Yu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Youhua Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
159
|
Scarfe L, Menshikh A, Newton E, Zhu Y, Delgado R, Finney C, de Caestecker MP. Long-term outcomes in mouse models of ischemia-reperfusion-induced acute kidney injury. Am J Physiol Renal Physiol 2019; 317:F1068-F1080. [PMID: 31411074 PMCID: PMC7132317 DOI: 10.1152/ajprenal.00305.2019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 07/31/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023] Open
Abstract
Severe acute kidney injury has a high mortality and is a risk factor for progressive chronic kidney disease. None of the potential therapies that have been identified in preclinical studies have successfully improved clinical outcomes. This failure is partly because animal models rarely reflect the complexity of human disease: most preclinical studies are short term and are commonly performed in healthy, young, male mice. Therapies that are effective in preclinical models that share common clinical features seen in patients with acute kidney injury, including genetic diversity, different sexes, and comorbidities, and evaluate long-term outcomes are more likely to predict success in the clinic. Here, we evaluated susceptibility to chronic kidney disease after ischemia-reperfusion injury with delayed nephrectomy by monitoring long-term functional and histological responses to injury. We defined conditions required to induce long-term postinjury renal dysfunction and fibrosis without increased mortality in a reproducible way and evaluate effect of mouse strains, sexes, and preexisting diabetes on these responses.
Collapse
Affiliation(s)
- Lauren Scarfe
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Anna Menshikh
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Emily Newton
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yuantee Zhu
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee
| | - Rachel Delgado
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Charlene Finney
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mark P de Caestecker
- Division of Nephrology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
160
|
Fu Y, Cai J, Li F, Liu Z, Shu S, Wang Y, Liu Y, Tang C, Dong Z. Chronic effects of repeated low-dose cisplatin treatment in mouse kidneys and renal tubular cells. Am J Physiol Renal Physiol 2019; 317:F1582-F1592. [PMID: 31532246 DOI: 10.1152/ajprenal.00385.2019] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cisplatin is a commonly used chemotherapeutic drug for cancer treatment, but its nephrotoxicity may lead to the deterioration of renal function. Previous work has been focused on cisplatin-induced acute kidney disease, whereas the mechanism of chronic kidney disease after cisplatin chemotherapy is largely unknown. In the present study, we have characterized the mouse model of chronic kidney defects induced by repeated low-dose cisplatin treatment. We have also established a relevant cell culture model. In the animal model, C57 mice were given weekly injection of 8 mg/kg cisplatin for 4 wk. This led to a sustained decline of kidney function. These mice showed loss of kidney mass, interstitial fibrosis, continued activation of inflammatory cytokines, and appearance of atubular glomeruli. In the cell model, the BUMPT mouse proximal tubular cell line was treated four times with 1-2 μM cisplatin, resulting in low levels of apoptosis and the expression of fibrosis proteins and profibrotic factors. These data suggest that repeated treatment with low-dose cisplatin causes long-term renal pathologies with characteristics of chronic kidney disease.
Collapse
Affiliation(s)
- Ying Fu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Juan Cai
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fanghua Li
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhiwen Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shaoqun Shu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ying Wang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuxue Liu
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chengyuan Tang
- Department of Nephrology, Hunan Key Laboratory of Kidney Disease and Blood Purification, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia
| |
Collapse
|
161
|
Silver SA, Gerarduzzi C. Found in Translation: Reasons for Optimism in the Pursuit to Prevent Chronic Kidney Disease After Acute Kidney Injury. Can J Kidney Health Dis 2019; 6:2054358119868740. [PMID: 31452903 PMCID: PMC6698989 DOI: 10.1177/2054358119868740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 06/21/2019] [Indexed: 12/14/2022] Open
Abstract
Purpose of review: The current review will discuss on the progress of studying the transition
phase between acute kidney injury (AKI) and chronic kidney disease (CKD)
through improved animal models, common AKI and CKD pathways, and how human
studies may inform different translational approaches. Sources of information: PubMed and Google Scholar. Methods: A narrative review was performed using the main terms “acute kidney injury,”
“chronic kidney disease,” “end-stage renal disease,” “animal models,”
“review,” “decision-making,” and “translational research.” Key findings: The last decade has shown much progress in the study of AKI, including
evidence of a pathophysiological link between AKI and CKD. We are now in a
phase of redesigning animal models and discovering mechanisms that can
replicate the pathological conditions of the AKI-to-CKD continuum.
Translating these findings into the clinic is a barrier that must be
overcome. To this end, current efforts include prediction of AKI onset and
maladaptive repair, detecting patients susceptible to the progression of
chronic maladaptive repair, and understanding shared signaling mechanisms
between AKI and CKD. Limitations: This is a narrative review of the literature that is partially influenced by
the knowledge, perspectives, and experiences of the authors and their
research background. Implications: Overall, this new knowledge from the AKI-to-CKD continuum will help bridge
the discontinuity that exists between animal models and patients, resulting
in more effective translational biomarkers and therapeutics to test in known
AKI pathologies thereby preventing the chronicity of kidney injury
progression.
Collapse
Affiliation(s)
- Samuel A. Silver
- Division of Nephrology, Kingston Health
Sciences Center, Queen’s University, Kingston, ON, Canada
| | - Casimiro Gerarduzzi
- Division de Néphrologie, Centre de
recherche de l’Hôpital Maisonneuve-Rosemont, Montréal, Québec, Canada
- Département de Médecine, Faculté de
Médecine, Université de Montréal, Montréal, Québec, Canada
- Casimiro Gerarduzzi, Division de
Néphrologie, Centre de recherche de l’Hôpital Maisonneuve-Rosemont, 5345,
boulevard de l’Assomption, Montreal, QC H1T 2M4, Canada.
| |
Collapse
|
162
|
Mechanisms of Fasting-Mediated Protection against Renal Injury and Fibrosis Development after Ischemic Acute Kidney Injury. Biomolecules 2019; 9:biom9090404. [PMID: 31443530 PMCID: PMC6770803 DOI: 10.3390/biom9090404] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 12/04/2022] Open
Abstract
Ischemia-reperfusion injury of the kidney may lead to renal fibrosis through a combination of several mechanisms. We recently demonstrated that fasting protects the rat kidney against oxidative stress and mitochondrial dysfunction in early acute kidney injury, and also against fibrosis development. Here we show that preoperative fasting preserves redox status and mitochondrial homeostasis at the chronic phase of damage after severe ischemia. Also, the protective effect of fasting coincides with the suppression of inflammation and endoplasmic reticulum stress, as well as the down-regulation of the mechanistic target of rapamycin (mTOR) and extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathways in the fibrotic kidney. Our results demonstrate that fasting targets multiple pathophysiological mechanisms to prevent renal fibrosis and damage that results after renal ischemia-reperfusion injury.
Collapse
|
163
|
Wei J, Zhang J, Wang L, Jiang S, Fu L, Buggs J, Liu R. New mouse model of chronic kidney disease transitioned from ischemic acute kidney injury. Am J Physiol Renal Physiol 2019; 317:F286-F295. [PMID: 31116604 PMCID: PMC6732455 DOI: 10.1152/ajprenal.00021.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 05/14/2019] [Accepted: 05/20/2019] [Indexed: 12/28/2022] Open
Abstract
Acute kidney injury (AKI) significantly increases the risk of development of chronic kidney disease (CKD), which is closely associated with the severity of AKI. However, the underlying mechanisms for the AKI to CKD transition remain unclear. Several animal models with AKI to CKD transition have been generated and widely used in research; however, none of them exhibit the typical changes in glomerular filtration rate or plasma creatinine, the hallmarks of CKD. In the present study, we developed a novel model with a typical phenotype of AKI to CKD transition in C57BL/6 mice. In this model, life-threatening ischemia-reperfusion injury was performed in one kidney, whereas the contralateral kidney was kept intact to maintain animal survival; then, after 2 wk of recovery, when the renal function of the injured kidney restored above the survival threshold, the contralateral intact kidney was removed. Animals of this two-stage unilateral ischemia-reperfusion injury model with pedicle clamping of 21 and 24 min exhibited an incomplete recovery from AKI and subsequent progression of CKD with characteristics of a progressive decline in glomerular filtration rate, increase in plasma creatinine, worsening of proteinuria, and deleterious histopathological changes, including interstitial fibrosis and glomerulosclerosis. In conclusion, a new model of the AKI to CKD transition was generated in C57BL/6 mice.
Collapse
Affiliation(s)
- Jin Wei
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| | - Jie Zhang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| | - Lei Wang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| | - Shan Jiang
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| | - Liying Fu
- Tampa General Hospital , Tampa, Florida
| | | | - Ruisheng Liu
- Department of Molecular Pharmacology and Physiology, University of South Florida College of Medicine , Tampa, Florida
| |
Collapse
|
164
|
Guan Y, Nakano D, Zhang Y, Li L, Tian Y, Nishiyama A. A mouse model of renal fibrosis to overcome the technical variability in ischaemia/reperfusion injury among operators. Sci Rep 2019; 9:10435. [PMID: 31320707 PMCID: PMC6639321 DOI: 10.1038/s41598-019-46994-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 07/08/2019] [Indexed: 12/16/2022] Open
Abstract
The ischaemia-reperfusion (I/R) model is a widely used model of acute kidney injury (AKI) and renal fibrosis. However, the ischaemia duration that is long enough to cause broad fibrosis shows that a high mortality rate and a short ischaemia duration does not cause fibrosis, resulting in a large variation in fibrosis progression in this experimental model. Inter-operator variation occurs for I/R injury severity because the I/R procedure is complex, which results in poor reproducibility of subsequent fibrosis in the model. In the present study, we developed a renal fibrosis model in which the fibrosis progression for 8 weeks is predictable within 8 days. Three operators independently performed I/R followed by uninephrectomy at day 7 in mice. The aim was to create a model that would show a blood urea nitrogen (BUN) level >100 mg/dL at day 8 after I/R (day 1 after uninephrectomy). Although the ischaemia duration to satisfy this BUN criterion differed among operators, the mice developed anaemia, polyuria, and fibrosis in a similar manner under the same BUN criterion with a low mortality rate. Interstitial fibrosis had developed at week 8, which was strongly correlated with the BUN at day 8. This protocol allows operators to adjust the ischaemia duration based on the BUN criterion and to separate mice into the desired number of groups based on the BUN to study interventions against renal fibrosis.
Collapse
Affiliation(s)
- Yu Guan
- Department of Pharmacology, Kagawa University Medical School, Kagawa, Japan.,Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Daisuke Nakano
- Department of Pharmacology, Kagawa University Medical School, Kagawa, Japan.
| | - Yifan Zhang
- Department of Pharmacology, Kagawa University Medical School, Kagawa, Japan.,Department of No.2 Orthopedics, Shijiazhuang City No.1 Hospital, Shijiazhuang, Hebei, China
| | - Lei Li
- Department of Pharmacology, Kagawa University Medical School, Kagawa, Japan
| | - Ye Tian
- Department of Urology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Akira Nishiyama
- Department of Pharmacology, Kagawa University Medical School, Kagawa, Japan
| |
Collapse
|
165
|
Perales-Quintana MM, Saucedo AL, Lucio-Gutiérrez JR, Waksman N, Alarcon-Galvan G, Govea-Torres G, Sanchez-Martinez C, Pérez-Rodríguez E, Guzman-de la Garza FJ, Cordero-Pérez P. Metabolomic and biochemical characterization of a new model of the transition of acute kidney injury to chronic kidney disease induced by folic acid. PeerJ 2019; 7:e7113. [PMID: 31275747 PMCID: PMC6590474 DOI: 10.7717/peerj.7113] [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: 01/30/2019] [Accepted: 05/10/2019] [Indexed: 12/24/2022] Open
Abstract
Background Renal diseases represent a major public health problem. The demonstration that maladaptive repair of acute kidney injury (AKI) can lead to the development of chronic kidney disease (CKD) and end-stage renal disease has generated interest in studying the pathophysiological pathways involved. Animal models of AKI–CKD transition represent important tools to study this pathology. We hypothesized that the administration of multiple doses of folic acid (FA) would lead to a progressive loss of renal function that could be characterized through biochemical parameters, histological classification and nuclear magnetic resonance (NMR) profiling. Methods Wistar rats were divided into groups: the control group received a daily intraperitoneal (I.P.) injection of double-distilled water, the experimental group received a daily I.P. injection of FA (250 mg kg body weight−1). Disease was classified according to blood urea nitrogen level: mild (40–80 mg dL−1), moderate (100–200 mg dL−1) and severe (>200 mg dL−1). We analyzed through biochemical parameters, histological classification and NMR profiling. Results Biochemical markers, pro-inflammatory cytokines and kidney injury biomarkers differed significantly (P < 0.05) between control and experimental groups. Histology revealed that as damage progressed, the degree of tubular injury increased, and the inflammatory infiltrate was more evident. NMR metabolomics and chemometrics revealed differences in urinary metabolites associated with CKD progression. The main physiological pathways affected were those involved in energy production and amino-acid metabolism, together with organic osmolytes. These data suggest that multiple administrations of FA induce a reproducible model of the induction of CKD. This model could help to evaluate new strategies for nephroprotection that could be applied in the clinic.
Collapse
Affiliation(s)
| | - Alma L Saucedo
- Analytic Chemistry Department, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | | | - Noemí Waksman
- Analytic Chemistry Department, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Gabriela Alarcon-Galvan
- Basic Science Department, School of Medicine, Universidad de Monterrey, Monterrey, Nuevo León, Mexico
| | - Gustavo Govea-Torres
- Liver Unit, Department of Internal Medicine, "Dr. José E. González" University Hospital, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Concepcion Sanchez-Martinez
- Nephrology Department, "Dr. José E. González" University Hospital, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | - Edelmiro Pérez-Rodríguez
- Transplant Service, "Dr. José E. González" University Hospital, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| | | | - Paula Cordero-Pérez
- Liver Unit, Department of Internal Medicine, "Dr. José E. González" University Hospital, Universidad Autónoma de Nuevo León, Monterrey, Nuevo León, Mexico
| |
Collapse
|
166
|
Soranno DE, Gil HW, Kirkbride-Romeo L, Altmann C, Montford JR, Yang H, Levine A, Buchanan J, Faubel S. Matching Human Unilateral AKI, a Reverse Translational Approach to Investigate Kidney Recovery after Ischemia. J Am Soc Nephrol 2019; 30:990-1005. [PMID: 31072827 DOI: 10.1681/asn.2018080808] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 03/14/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The duration of renal ischemia that is associated with (or leads to) renal injury in patients is uncertain, and a reverse translational research approach has been proposed to improve animal models of AKI to facilitate clinical translatability. We developed a two murine models of unilateral renal ischemia to match a recently published human study that investigated renal injury after unilateral renal ischemia during partial nephrectomy. METHODS Eight 10-week-old C57BL/6 male mice underwent left UiAKI or sham procedure, with or without intra-operative ice packs. Functional, histological, and biomarker outcomes were followed at 2, 6 and 24 hours, or 14 or 28 days later. The 14 and 28 day cohorts were duplicated such that contralateral nephrectomy could be performed 3 days prior to sacrifice with functional measurements obtained to isolate the glomerular filtration rate of the injured kidney. RESULTS The short-term outcomes correlated with the human study findings with urine and serum biomarkers of injury peaking around 24 hours and then normalizing, and reassuring immediate histological outcomes. Functional and histological outcomes at the later time-points (14 and 28 days) demonstrate an increase in fibrosis markers, and a reduction in glomerular filtration rate in the injured kidney, corresponding to the duration of ischemia, while serum and urine biomarkers remained reassuring. CONCLUSIONS Our findings suggest that clinically available biomarkers of renal function are falsely reassuring against long-term injury following UiAKI, and that the duration of ischemia correlates with impaired function and increased fibrosis.
Collapse
Affiliation(s)
- Danielle E Soranno
- Division of Renal Disease and Hypertension, Department of Medicine.,Section of Pediatric Nephrology, Department of Pediatrics, and.,Department of Bioengineering, University of Colorado, Aurora, Colorado
| | - Hyo-Wook Gil
- Division of Renal Disease and Hypertension, Department of Medicine.,Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, Cheonan, Korea
| | | | | | - John R Montford
- Division of Renal Disease and Hypertension, Department of Medicine.,Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, Colorado; and
| | - Haichun Yang
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ani Levine
- Department of Bioengineering, University of Colorado, Aurora, Colorado
| | - Jane Buchanan
- Section of Pediatric Nephrology, Department of Pediatrics, and
| | - Sarah Faubel
- Division of Renal Disease and Hypertension, Department of Medicine, .,Section of Pediatric Nephrology, Department of Pediatrics, and
| |
Collapse
|
167
|
Manolopoulou M, Matlock BK, Nlandu-Khodo S, Simmons AJ, Lau KS, Phillips-Mignemi M, Ivanova A, Alford CE, Flaherty DK, Gewin LS. Novel kidney dissociation protocol and image-based flow cytometry facilitate improved analysis of injured proximal tubules. Am J Physiol Renal Physiol 2019; 316:F847-F855. [PMID: 30759021 PMCID: PMC6580245 DOI: 10.1152/ajprenal.00354.2018] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 02/08/2019] [Accepted: 02/11/2019] [Indexed: 12/16/2022] Open
Abstract
Flow cytometry studies on injured kidney tubules are complicated by the low yield of nucleated single cells. Furthermore, cell-specific responses such as cell cycle dynamics in vivo have conventionally relied on indirect immunohistochemistry and proximal tubule markers that may be downregulated in injury. Here, we report a new tissue dissociation protocol for the kidney with an early fixation step that greatly enhances the yield of single cells. Genetic labeling of the proximal tubule with either mT/mG "tomato" or R26Fucci2aR (Fucci) cell cycle reporter mice allows us to follow proximal tubule-specific changes in cell cycle after renal injury. Image-based flow cytometry (FlowSight) enables gating of the cell cycle and concurrent visualization of the cells with bright field and fluorescence. We used the Fucci mouse in conjunction with FlowSight to identify a discrete polyploid population in proximal tubules after aristolochic acid injury. The tissue dissociation protocol in conjunction with genetic labeling and image-based flow cytometry is a tool that can improve our understanding of any discrete cell population after injury.
Collapse
Affiliation(s)
- Marika Manolopoulou
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Brittany K Matlock
- Flow Cytometry Shared Resource, Vanderbilt Vaccine Center, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Stellor Nlandu-Khodo
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Alan J Simmons
- Epithelial Biology Center, Vanderbilt University Medical Center , Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University , Nashville, Tennessee
| | - Ken S Lau
- Epithelial Biology Center, Vanderbilt University Medical Center , Nashville, Tennessee
- Center for Quantitative Sciences, Vanderbilt University Medical Center , Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University , Nashville, Tennessee
- Program in Chemical and Physical Biology, Vanderbilt University , Nashville, Tennessee
| | - Melanie Phillips-Mignemi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Alla Ivanova
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Catherine E Alford
- Department of Pathology and Laboratory Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
| | - David K Flaherty
- Flow Cytometry Shared Resource, Vanderbilt Vaccine Center, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Leslie S Gewin
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee
- Department of Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University , Nashville, Tennessee
| |
Collapse
|
168
|
Xu L, Li X, Zhang F, Wu L, Dong Z, Zhang D. EGFR drives the progression of AKI to CKD through HIPK2 overexpression. Theranostics 2019; 9:2712-2726. [PMID: 31131063 PMCID: PMC6526000 DOI: 10.7150/thno.31424] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/25/2019] [Indexed: 12/30/2022] Open
Abstract
The molecular mechanism underlying the transition of acute kidney injury (AKI) to chronic kidney disease (CKD) induced by vancomycin (VAN) remains largely unknown. Methods: The mice model of VAN drives AKI to CKD was developed to investigate the role and molecular mechanism of epidermal growth factor receptor (EGFR). The EGF receptor mutant (Wa-2) mice and gefitinib were used to inactivation of EGFR. The homeodomain interacting protein kinase 2 (HIPK2) siRNA was applied to silence of HIPK2. Human proximal tubular epithelial cells (HK-2) were used to explore the molecular regulation methanism of EGFR. ChIp analysis was used to investigate if STAT3 interaction with the promoter of HIPK2. Results: A novel VAN-induced AKI mouse model was established for the first time. Moreover, the expression levels collagen I&IV, α-SMA, p-EGFR and the expression of HIPK2 proteins were upregulated in this model. Interestingly, AKI caused by VAN was markedly attenuated in waved-2 mice at the early stage, as evidenced by the suppression of renal dysfunction, renal cell apoptosis and caspase3 activation. In the latter stage, renal fibrosis and inflammation were significantly ameliorated in Wa-2 mice, accompanied by the downregulation of profibrotic molecules and F4/80. Besides, the expression levels of HIPK2 and p-STAT3 were suppressed in Wa-2 mice during VAN-induced transition of AKI to CKD. In addition, renal fibrosis and inflammation, profibrotic molecules, and EGFR/STAT3/HIPK2 signaling were ameliorated by gefitinib treatment after VAN-induced AKI. These results were consistent with the findings of Wa-2 mice. EGFR/STAT3 signaling mediated VAN-induced HIPK2 expression in HK-2 cells. ChIp analysis revealed that STAT3 directly bound to the promoter region of HIPK2. Finally, inhibition of HIPK2 attenuated the VAN drove the progression of AKI to CKD. Conclusion: These data suggest that EGFR plays an important role in VAN-driven progression of AKI to CKD.
Collapse
MESH Headings
- Actins/genetics
- Actins/metabolism
- Acute Kidney Injury/chemically induced
- Acute Kidney Injury/complications
- Acute Kidney Injury/genetics
- Acute Kidney Injury/metabolism
- Animals
- Calcium-Binding Proteins/genetics
- Calcium-Binding Proteins/metabolism
- Caspase 3/genetics
- Caspase 3/metabolism
- Cell Line
- Collagen Type I/genetics
- Collagen Type I/metabolism
- Collagen Type IV/genetics
- Collagen Type IV/metabolism
- Epithelial Cells/cytology
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Gefitinib/pharmacology
- Gene Expression Regulation
- Humans
- Kidney/drug effects
- Kidney/metabolism
- Kidney/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Mutation
- Promoter Regions, Genetic
- Protein Binding
- Protein Kinase Inhibitors/pharmacology
- Protein Serine-Threonine Kinases/antagonists & inhibitors
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Renal Insufficiency, Chronic/chemically induced
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/metabolism
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Signal Transduction
- Vancomycin/administration & dosage
Collapse
Affiliation(s)
- Luyang Xu
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, Hunan, People's Republic of China
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China, People's Republic of China
| | - Xiaozhou Li
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, Hunan, People's Republic of China
| | - Fei Zhang
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, Hunan, People's Republic of China
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China, People's Republic of China
| | - Lidong Wu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China, People's Republic of China
| | - Zheng Dong
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Department of cellular Biology and anatomy, Medical college of Georgia at Georgia Regents University and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Dongshan Zhang
- Department of Emergency Medicine, Second Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Emergency Medicine and Difficult Diseases Institute, Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
169
|
Baseline Serum Cystatin C Is a Potential Predictor for Acute Kidney Injury in Patients with Acute Pancreatitis. DISEASE MARKERS 2018; 2018:8431219. [PMID: 30581500 PMCID: PMC6276423 DOI: 10.1155/2018/8431219] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 09/04/2018] [Accepted: 09/20/2018] [Indexed: 02/08/2023]
Abstract
Aims The study is aimed at studying the incidence of acute kidney injury (AKI) and exploring the potential predictor for AKI in patients with acute pancreatitis. Methods A retrospective study adopting a stratified cohort sampling design was performed in a cohort of patients (n = 237) diagnosed with acute pancreatitis without any renal injury. The following information including age, gender, serum creatinine, serum urea nitrogen, serum uric acid, serum cystatin C, fasting serum glucose, serum amylase, serum lipase, serum choline esterase, total protein, albumin, globulin, total bilirubin, direct bilirubin, total bile acids, glutamic-pyruvic transaminase, glutamic-oxaloacetic transaminase, gamma glutamyl transpeptidase, and alkaline phosphatase were collected from each patient when they were diagnosed with acute pancreatitis. Student t-test was conducted to figure out the difference between patients with and without AKI. Univariate and multivariate logistic regression analyses were used for investigating the predictors for AKI in patients with acute pancreatitis. Results 18 (7.6%) patients in total had developed AKI among the study group. Compared with patients without AKI (1.01 ± 0.26 mg/L), the level of baseline serum cystatin C (CYS-C) was significantly higher in patients with AKI (3.64 ± 2.17 mg/L, P < 0.001). Baseline serum CYS-C (OR = 203.594, P < 0.001) was the independent and significant predictor for AKI in patients with acute pancreatitis. AKI in patients with acute pancreatitis could be identified with a sensitivity of 88.9% at specificity of 100% (AUC = 0.948, 95% CI 0.879–1.000) by baseline serum CYS-C (cut-off value = 1.865 mg/L). Conclusions Baseline serum CYS-C shall be adopted to predict the potential risk of AKI in patients with acute pancreatitis.
Collapse
|
170
|
Shu S, Zhu J, Liu Z, Tang C, Cai J, Dong Z. Endoplasmic reticulum stress is activated in post-ischemic kidneys to promote chronic kidney disease. EBioMedicine 2018; 37:269-280. [PMID: 30314894 PMCID: PMC6286638 DOI: 10.1016/j.ebiom.2018.10.006] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 09/22/2018] [Accepted: 10/02/2018] [Indexed: 02/06/2023] Open
Abstract
Background Acute kidney injury (AKI) may lead to the development of chronic kidney disease (CKD), i.e. AKI-CKD transition, but the underlying mechanism remains largely unclear. Endoplasmic reticulum (ER) stress is characterized by the accumulation of unfolded or misfolded proteins in ER resulting in a cellular stress response. The role of ER stress in AKI-CKD transition remains unknown. Methods In this study, we examined ER stress in the mouse model of AKI-CKD transition after unilateral renal ischemia-reperfusion injury (uIR). To determine the role of ER stress in AKI-CKD transition, we tested the effects of two chemical chaperones: Tauroursodeoxycholic acid (TUDCA) and 4-phenylbutyric acid (4-PBA). Findings uIR led to the induction of ER stress in kidneys, as indicated by increased expression of UPR molecules CHOP (C/EBP homologous protein) and BiP(binding immunoglobulin protein; also called GRP78–78 kDa glucoseregulated protein). Given at 3 days after uIR, both TUDCA and 4-PBA blocked ER stress in post-ischemic kidneys. Notably, both chemicals promoted renal recovery and suppressed tubulointerstitial injury as manifested by the reduction of tubular atrophy, renal fibrosis and myofibroblast activation. Inhibition of ER stress further attenuated renal tubular epithelial cell apoptosis, inflammation and autophagy in post-ischemic kidneys. Interpretation These findings suggest that ER stress contributes critically to the development of chronic kidney pathologies and CKD following AKI, and inhibition of ER stress may represent a potential therapeutic strategy to impede AKI-CKD transition.
Collapse
Affiliation(s)
- Shaoqun Shu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Jiefu Zhu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Zhiwen Liu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Chengyuan Tang
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Juan Cai
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Changsha, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
171
|
Polichnowski AJ. Microvascular rarefaction and hypertension in the impaired recovery and progression of kidney disease following AKI in preexisting CKD states. Am J Physiol Renal Physiol 2018; 315:F1513-F1518. [PMID: 30256130 DOI: 10.1152/ajprenal.00419.2018] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute kidney injury (AKI) is a major complication in hospitalized patients and is associated with elevated mortality rates. Numerous recent studies indicate that AKI also significantly increases the risk of chronic kidney disease (CKD), end-stage renal disease (ESRD), hypertension, cardiovascular disease, and mortality in those patients who survive AKI. Moreover, the risk of ESRD and mortality after AKI is substantially higher in patients with preexisting CKD. However, the underlying mechanisms by which AKI and CKD interact to promote ESRD remain poorly understood. The recently developed models that superimpose AKI on rodents with preexisting CKD have provided new insights into the pathogenic mechanisms mediating the deleterious interactions between AKI and CKD. These studies show that preexisting CKD impairs recovery from AKI and promotes the development of mechanisms of CKD progression. Specifically, preexisting CKD exacerbates microvascular rarefaction, failed tubular redifferentiation, disruption of cell cycle regulation, hypertension, and proteinuria after AKI. The purpose of this review is to discuss the potential mechanisms by which microvascular rarefaction and hypertension contribute to impaired recovery from AKI and the subsequent progression of renal disease in preexisting CKD states.
Collapse
Affiliation(s)
- Aaron J Polichnowski
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee.,Center of Excellence in Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee
| |
Collapse
|