1
|
Sadeghmanesh F, Eidi A, Mortazavi P, Oryan S. Nanoselenium attenuates renal ischemia-reperfusion injury in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2297-2310. [PMID: 37819388 DOI: 10.1007/s00210-023-02723-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/13/2023] [Indexed: 10/13/2023]
Abstract
Using selenium (Se) nanoparticles has received attention in recent years because of their therapeutic benefits due to their anticancer, antioxidant, anti-inflammatory, and anti-diabetic effects. This research was conducted to evaluate the possible protective impact of nano-Se on renal unilateral ischemia/reperfusion injury (uIRI) in adult male Wistar rats. Using clamping of the left renal pedicle within 45 min uIRI was induced. The animals were randomly divided into nine groups of control, nano-Se (0.25, 0.5, and 1 mg/kg bw/day) alone, uIRI control, and uIRI rats administrated with nano-Se. At 30 days after treatment, the animals were sacrificed to be assessed biochemically and histopathologically. Nano-Se in uIRI groups have significantly decreased serum creatinine, urea levels, renal histological damage, and increased antioxidant status. Also, our findings demonstrated that the administration of nano-Se caused a significant decrease in the immunoreactivity level of the epidermal growth factor (EGF) and EGFR expression (EGF receptor) in the renal tissue of the uIRI rats. Therefore, nano-Se possesses renoprotective effects, and this effect might be attributable to its antioxidant and free radical scavenger effects. These renoprotective effects may depend on the decreased EGF immunoreactivity level and EGFR expression in the kidney tissue and improve the structure of the kidney tissue. Thus, our research provided biochemical and histological data supporting the potential clinical use of nano-Se for the treatment of certain kidney disorders.
Collapse
Affiliation(s)
- Farzaneh Sadeghmanesh
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Akram Eidi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Pejman Mortazavi
- Department of Pathology, Faculty of Specialized Veterinary Sciences, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | |
Collapse
|
2
|
Shao L, Fang Q, Shi C, Zhang Y, Xia C, Zhang Y, Wang J, Chen F. Bone marrow mesenchymal stem cells inhibit ferroptosis via regulating the Nrf2-keap1/p53 pathway to ameliorate chronic kidney disease injury in the rats. J Recept Signal Transduct Res 2023; 43:9-18. [PMID: 36883687 DOI: 10.1080/10799893.2023.2185083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
PURPOSE Although bone marrow mesenchymal stem cells (BMMSCs) have been reported to exhibit a protective effect on animal models of chronic kidney disease (CKD), the exact mechanisms involved require further investigation. This study aims to investigate the underlying molecular mechanisms of BMMSCs in inhibiting ferroptosis and preventing an Adriamycin (ADR)-induced CKD injury. METHODS A rat model of long-term CKD induced through the injection of ADR administered twice weekly via the tail vein was used in this study. After BMMSCs were systemically administered through the renal artery, pathological staining, western blotting, ELISA, and transmission electron microscopy were used to analyze ferroptosis. RESULTS Analyses of renal function and histopathological findings indicated that ADR-mediated renal dysfunction improved in response to the BMMSC treatment, which was also sufficient to mediate the partial reversal of renal injury and mitochondrial pathological changes. BMMSCs decreased the ferrous iron (Fe2+) and reactive oxygen species and elevated glutathione (GSH) and GSH peroxidase 4. Moreover, the BMMSC treatment activated the expression of ferroptosis-related regulator NF-E2-related factor 2 (Nrf2) and inhibited Keap1 and p53 in CKD rat kidney tissues. CONCLUSIONS BMMSCs alleviate CKD, possibly resulting from the inhibition of kidney ferroptosis by regulating the Nrf2-Keap1/p53 pathway.
Collapse
Affiliation(s)
- Lishi Shao
- Department of Radiology, Kunming Medical University & The Second Affiliated Hospital, Kunming, Yunnan
| | - Qixiang Fang
- Department of Urology, The First Affiliated Hospital of the Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi
| | - Chen Shi
- Department of Radiology, Kunming Medical University & The Second Affiliated Hospital, Kunming, Yunnan
| | - Ya Zhang
- Department of Radiology, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan
| | - Chunjuan Xia
- Department of Radiology, Kunming Medical University & The Second Affiliated Hospital, Kunming, Yunnan
| | - Yifan Zhang
- Department of Radiology, Kunming Medical University & The Second Affiliated Hospital, Kunming, Yunnan
| | - Jiaping Wang
- Department of Radiology, Kunming Medical University & The Second Affiliated Hospital, Kunming, Yunnan
| | - Fukun Chen
- Department of Nuclear Medicine, Yunnan Cancer Hospital & The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan
| |
Collapse
|
3
|
Voggel J, Mohr J, Nüsken KD, Dötsch J, Nüsken E, Alejandre Alcazar MA. Translational insights into mechanisms and preventive strategies after renal injury in neonates. Semin Fetal Neonatal Med 2022; 27:101245. [PMID: 33994314 DOI: 10.1016/j.siny.2021.101245] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Adverse perinatal circumstances can cause acute kidney injury (AKI) and contribute to chronic kidney disease (CKD). Accumulating evidence indicate that a wide spectrum of perinatal conditions interferes with normal kidney development and ultimately leads to aberrant kidney structure and function later in life. The present review addresses the lack of mechanistic knowledge with regard to perinatal origins of CKD and provides a comprehensive overview of pre- and peri-natal insults, including genetic predisposition, suboptimal nutritional supply, obesity and maternal metabolic disorders as well as placental insufficiency leading to intrauterine growth restriction (IUGR), prematurity, infections, inflammatory processes, and the need for life-saving treatments (e.g. oxygen supplementation, mechanical ventilation, medications) in neonates. Finally, we discuss future preventive, therapeutic, and regenerative directions. In summary, this review highlights the perinatal vulnerability of the kidney and the early origins of increased susceptibility toward AKI and CKD during postnatal life. Promotion of kidney health and prevention of disease require the understanding of perinatal injury in order to optimize perinatal micro- and macro-environments and enable normal kidney development.
Collapse
Affiliation(s)
- Jenny Voggel
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Germany
| | - Jasmine Mohr
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Germany
| | - Kai-Dietrich Nüsken
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany
| | - Jörg Dötsch
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany
| | - Eva Nüsken
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Department of Pediatric and Adolescent Medicine, Germany
| | - Miguel A Alejandre Alcazar
- University of Cologne, Faculty of Medicine, University Hospital Cologne, Translational Experimental Pediatrics - Experimental Pulmonology, Department of Pediatric and Adolescent Medicine, Germany; University of Cologne, Faculty of Medicine, University Hospital Cologne, Center for Molecular Medicine Cologne (CMMC), Germany; Excellence Cluster on Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Faculty of Medicine, University Hospital Cologne Cologne, Germany; Institute for Lung Health, University of Giessen and Marburg Lung Centre (UGMLC), Member of the German Centre for Lung Research (DZL), Gießen, Germany.
| |
Collapse
|
4
|
Caneparo C, Sorroza-Martinez L, Chabaud S, Fradette J, Bolduc S. Considerations for the clinical use of stem cells in genitourinary regenerative medicine. World J Stem Cells 2021; 13:1480-1512. [PMID: 34786154 PMCID: PMC8567446 DOI: 10.4252/wjsc.v13.i10.1480] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/12/2021] [Accepted: 09/17/2021] [Indexed: 02/06/2023] Open
Abstract
The genitourinary tract can be affected by several pathologies which require repair or replacement to recover biological functions. Current therapeutic strategies are challenged by a growing shortage of adequate tissues. Therefore, new options must be considered for the treatment of patients, with the use of stem cells (SCs) being attractive. Two different strategies can be derived from stem cell use: Cell therapy and tissue therapy, mainly through tissue engineering. The recent advances using these approaches are described in this review, with a focus on stromal/mesenchymal cells found in adipose tissue. Indeed, the accessibility, high yield at harvest as well as anti-fibrotic, immunomodulatory and proangiogenic properties make adipose-derived stromal/SCs promising alternatives to the therapies currently offered to patients. Finally, an innovative technique allowing tissue reconstruction without exogenous material, the self-assembly approach, will be presented. Despite advances, more studies are needed to translate such approaches from the bench to clinics in urology. For the 21st century, cell and tissue therapies based on SCs are certainly the future of genitourinary regenerative medicine.
Collapse
Affiliation(s)
- Christophe Caneparo
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
| | - Luis Sorroza-Martinez
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
| | - Stéphane Chabaud
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
| | - Julie Fradette
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec G1V0A6, Canada
| | - Stéphane Bolduc
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Quebec G1J1Z4, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec G1V0A6, Canada
| |
Collapse
|
5
|
Myla A, Dasmahapatra AK, Tchounwou PB. Sex-reversal and Histopathological Assessment of Potential Endocrine-Disrupting Effects of Graphene Oxide on Japanese medaka (Oryzias latipes) Larvae. CHEMOSPHERE 2021; 279:130768. [PMID: 34134430 PMCID: PMC8217731 DOI: 10.1016/j.chemosphere.2021.130768] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/24/2021] [Accepted: 04/28/2021] [Indexed: 05/12/2023]
Abstract
Sex-ratio is considered as an end point during endocrine disrupting chemicals (EDCs) evaluation. Many fish species including Japanese medaka have XX/XY sex determination mechanism, however, sex reversal (SR) can be induced by external and genetic factors. SR imposed an imbalance in natural sex ratio of a population living in any ecosystem. Considering SR as an end point, we aimed to investigate the potential EDC effects of graphene oxide (GO), a nanocarbon, using Japanese medaka as a model. One-day post-hatch (dph) medaka fries were exposed to GO (2.5, 5.0, 10.0 and 20 mg/L) for 96 h without food, followed by 6 weeks depuration in a GO-free environment with feeding. Phenotypic sex was determined by gonad histology; genotypic sex by genotyping Y-chromosome-specific male sex determining gene, dmy. Our data indicated testes in both XY and XX genotypes, while ovaries were only in XX females. Histopathology of XY and XX testis showed isogenic spermatocysts with active spermatogenesis. Distribution of spermatocytes (SPTs), not the spermatogonium (SPGs), showed enhancement in XY than XX testis. Female phenotypes had single ovary, either in stage 0 or 1. Ovo-testis/testis-ova were absent in XX or XY gonads. GO (2.5-20 mg/L) had inconsistent concentration-dependent effect in both SPGs and SPTs; however, no effect on ovarian follicles. Despite genotypic differences (XY/XX), in the histopathology/histochemistry of liver and kidneys GO effects was found to be minimum. Taken together, present study showed spontaneous induction of SR in some XX genotypes; however, exposure of fasting fries to GO had no apparent EDC effects.
Collapse
Affiliation(s)
- Anitha Myla
- RCMI Center for Environmental Health, Jackson State University, Jackson, MS, 39217, USA
| | - Asok K Dasmahapatra
- RCMI Center for Environmental Health, Jackson State University, Jackson, MS, 39217, USA; Department of BioMolecular Sciences, Environmental Toxicology Division, University of Mississippi, University, MS, 38677, USA
| | - Paul B Tchounwou
- RCMI Center for Environmental Health, Jackson State University, Jackson, MS, 39217, USA.
| |
Collapse
|
6
|
Borges FT, Convento MB, Schor N. Bone marrow-derived mesenchymal stromal cell: what next? STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2018; 11:77-83. [PMID: 30510433 PMCID: PMC6231430 DOI: 10.2147/sccaa.s147804] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Bone marrow mesenchymal stromal cell (MSC) is a potential alternative in regenerative medicine and has great potential in many pathologic conditions including kidney disease. Although most of the studies demonstrate MSC efficiency, the regenerative potential may not be efficient in all diseases and patients. Stem cell feasibility is modified by donor characteristics as gender, age, diet, and health status, producing both positive and negative results. The conditioning of MSC can potentiate its effects and modify its culture medium (CM). In current practices, the cell-free treatment is gaining notable attention, while MSC-conditioned CM is being applied and studied in many experimental diseases, including, but not limited to, certain kidney diseases. This may be the next step for clinical trials. Studies in stem cell CM have focused mainly on extracellular vesicles, nucleic acids (mRNA and microRNA), lipids, and proteins presented in this CM. They mediate regenerative effects of MSC in a harmonic manner. In this review, we will analyze the regenerative potential of MSC and its CM as well as discuss some effective techniques for modifying its fractions and improving its therapeutic potential. CM fractions may be modified by hypoxic conditions, inflammation, lipid exposition, and protein growth factors. Other possible mechanisms of action of stem cells are also suggested. In the future, the MSC paracrine effect may be modified to more closely meet each patient’s needs.
Collapse
Affiliation(s)
- Fernanda T Borges
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo, SP, Brazil, .,Interdisciplinary Postgraduate Program in Health Sciences, Universidade Cruzeiro do Sul, São Paulo, SP, Brazil,
| | - Marcia Bastos Convento
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo, SP, Brazil,
| | - Nestor Schor
- Nephrology Division, Department of Medicine, Universidade Federal de São Paulo, São Paulo, SP, Brazil,
| |
Collapse
|
7
|
Fan M, Zhang J, Xin H, He X, Zhang X. Current Perspectives on Role of MSC in Renal Pathophysiology. Front Physiol 2018; 9:1323. [PMID: 30294285 PMCID: PMC6158317 DOI: 10.3389/fphys.2018.01323] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 08/31/2018] [Indexed: 01/06/2023] Open
Abstract
In the course of the development and worsening of kidney disease, the treatments available are expensive and may cause adverse effects such as immune rejection, inadequate renal resources, or post-operative complications. Therefore, there is an urgent to develop more effective treatments. The advent of mesenchymal stem cells (MSCs) represents a new direction in this context. The current use of MSCs for the treatment of kidney disease has mostly involved experimental studies on animals and only a few clinical trials have been conducted. This review focused on the mechanisms of MSC involvement from different sources in the improvement of renal pathophysiology in recent years. These mechanisms include homing to damaged kidney tissue, and differentiating into or fusing with the innate cells of the kidney. The paracrine or endocrine action through secreting protective cytokines and/or releasing microvesicle from MSCs also plays a critical role in amelioration of kidney disease. With modern engineering technology like microRNA delivery and a combinational therapy approach such as reduction of renal fibrosis in obstructive nephropathy with MSCs and serelaxin, MSC may make great contribution to the improvement of renal pathophysiology. However, the therapeutic effects of MSC are still controversial and several problems remain unsolved. While it is too early to state that MSCs are useful for the treatment of renal diseases in clinic, it is thought that solutions to the existing problems will enable effective modulation of the biological characteristics of MSCs, thereby providing new and effective approaches for the treatment of renal diseases.
Collapse
Affiliation(s)
- Min Fan
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jing Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Hong Xin
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xiaozhou He
- The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Regenerative medicine in kidney disease: where we stand and where to go. Pediatr Nephrol 2018; 33:1457-1465. [PMID: 28735502 DOI: 10.1007/s00467-017-3754-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 05/23/2017] [Accepted: 05/26/2017] [Indexed: 02/07/2023]
Abstract
The kidney is a complex organ with more than 20 types of specialized cells that play an important role in maintaining the body's homeostasis. The epithelial tubular cell is formed during embryonic development and has little proliferative capacity under physiological conditions, but after acute injury the kidney does have regenerative capacity. However, after repetitive or severe lesions, it may undergo a maladaptation process that predisposes it to chronic kidney injury. Regenerative medicine includes various repair and regeneration techniques, and these have gained increasing attention in the scientific literature. In the future, not only will these techniques contribute to the repair and regeneration of the human kidney, but probably also to the construction of an entire organ. New mechanisms studied for kidney regeneration and repair include circulating stem cells as mesenchymal stromal/stem cells and their paracrine mechanisms of action; renal progenitor stem cells; the leading role of tubular epithelial cells in the tubular repair process; the study of zebrafish larvae to understand the process of nephron development, kidney scaffold and its repopulation; and, finally, the development of organoids. This review elucidates where we are in terms of current scientific knowledge regarding these mechanisms and the promises of future scientific perspectives.
Collapse
|
9
|
Renoprotective Effect of Platelet-Rich Plasma on Cisplatin-Induced Nephrotoxicity in Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9658230. [PMID: 30116500 PMCID: PMC6079401 DOI: 10.1155/2018/9658230] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 06/27/2018] [Indexed: 12/31/2022]
Abstract
Platelet-rich plasma (PRP) has grown as an attractive biologic instrument in regenerative medicine for its powerful healing properties. It is considered as a source of growth factors that may induce tissue repairing and improve fibrosis. This product has proven its efficacy in multiple studies, but its effect on cisplatin-induced nephrotoxicity has not yet been elucidated. The present investigation was performed to estimate the protective impact of platelet-rich plasma against cisplatin- (CP-) evoked nephrotoxicity in male rats. Nephrotoxicity was induced in male Wistar rats by right uninephrectomy followed by CP administration. Uninephrectomized rats were assigned into four groups: (1) control group, (2) PRP group, (3) CP group, and (4) CP + PRP group. PRP was administered by subcapsular renal injection. Renal function, inflammatory cytokines, and growth factor level as well as histopathological investigation were carried out. Treatment with PRP attenuated the severity of CP-induced nephrotoxicity as evidenced by suppressed creatinine, blood urea nitrogen (BUN), and N-acetyl glucosaminidase (NAG) levels. Moreover, PRP depressed intercellular adhesion molecule-1 (ICAM-1), kidney injury molecule-1 (KIM-1), caspase-3, and transforming growth factor-beta 1 (TGF-β1) levels, while enhanced the epidermal growth factor (EGF) level. These biochemical results were reinforced by the histopathological investigation, which revealed restoration of normal renal tissue architectures. These findings highlight evidence for the possible protective effects of PRP in a rat model of CP-induced nephrotoxicity, suggesting a new avenue for using PRP to improve the therapeutic index of cisplatin.
Collapse
|
10
|
He L, Wei Q, Liu J, Yi M, Liu Y, Liu H, Sun L, Peng Y, Liu F, Venkatachalam MA, Dong Z. AKI on CKD: heightened injury, suppressed repair, and the underlying mechanisms. Kidney Int 2017; 92:1071-1083. [PMID: 28890325 DOI: 10.1016/j.kint.2017.06.030] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 06/13/2017] [Accepted: 06/19/2017] [Indexed: 02/07/2023]
Abstract
Acute kidney injury (AKI) and chronic kidney disease (CKD) are interconnected. Although AKI-to-CKD transition has been intensively studied, the information of AKI on CKD is very limited. Nonetheless, AKI, when occurring in patients with CKD, is known to be more severe and difficult to recover. CKD is associated with significant changes in cell signaling in kidney tissues, including the activation of transforming growth factor-β, p53, hypoxia-inducible factor, and major developmental pathways. At the cellular level, CKD is characterized by mitochondrial dysfunction, oxidative stress, and aberrant autophagy. At the tissue level, CKD is characterized by chronic inflammation and vascular dysfunction. These pathologic changes may contribute to the heightened sensitivity of, and nonrecovery from, AKI in patients with CKD.
Collapse
Affiliation(s)
- Liyu He
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Qingqing Wei
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, Georgia, USA
| | - Jing Liu
- Department of Nephrology, The Second Xiangya Hospital, 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, Georgia, USA
| | - Mixuan Yi
- Department of Nephrology, The Second Xiangya Hospital, 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, Georgia, USA
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hong Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Sun
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Youming Peng
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fuyou Liu
- Department of Nephrology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Manjeri A Venkatachalam
- Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA
| | - Zheng Dong
- Department of Nephrology, The Second Xiangya Hospital, 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, Georgia, USA.
| |
Collapse
|
11
|
Kaur A, Sharma S. Mammalian target of rapamycin (mTOR) as a potential therapeutic target in various diseases. Inflammopharmacology 2017; 25:293-312. [PMID: 28417246 DOI: 10.1007/s10787-017-0336-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/02/2017] [Indexed: 12/28/2022]
Abstract
Mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase that belongs to Phosphatidylinositol-3-kinase related kinase superfamily. The signaling pathways of mTOR are integrated through the protein complexes of mTORC1 and mTORC2. mTORC1 controls protein synthesis, cell growth, proliferation, autophagy, cell metabolism, and stress responses, whereas mTORC2 seems to regulate cell survival and polarity. Dysregulation of the mTOR pathway has been implicated in the pathophysiology of a number of disease conditions, including cancer, cardiovascular, neurodegenerative, and various renal diseases. The hyperactivation of the mTOR pathway leads to increase in cell growth and proliferation and also has been documented to stimulate tumor growth. Therefore, investigation of the involvement of mTOR and its downstream pathways in various diseases intensively preoccupied scientific community. The present review is focussed on recent advances in the understanding of the mTOR signaling pathway and its role in health and various diseases.
Collapse
Affiliation(s)
- Avileen Kaur
- Cardiovascular Division, Department of Pharmacology, I. S. F. College of Pharmacy, Moga, Punjab, 142001, India
| | - Saurabh Sharma
- Cardiovascular Division, Department of Pharmacology, I. S. F. College of Pharmacy, Moga, Punjab, 142001, India.
| |
Collapse
|
12
|
Effect of Chinese Materia Medica with Tonifying Kidney Function on Transplantation of Multipotency Mesenchymal Stem Cells from Human Umbilical Cord in Mice Model of Acute Kidney Injury. CHINESE HERBAL MEDICINES 2016. [DOI: 10.1016/s1674-6384(16)60027-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
|
13
|
Stem cells and kidney regeneration. J Formos Med Assoc 2014; 113:201-9. [PMID: 24434243 DOI: 10.1016/j.jfma.2013.12.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Revised: 11/13/2013] [Accepted: 12/09/2013] [Indexed: 12/24/2022] Open
Abstract
Kidney disease is an escalating burden all over the world. In addition to preventing kidney injury, regenerating damaged renal tissue is as important as to retard the progression of chronic kidney disease to end stage renal disease. Although the kidney is a delicate organ and has only limited regenerative capacity compared to the other organs, an increasing understanding of renal development and renal reprogramming has kindled the prospects of regenerative options for kidney disease. Here, we will review the advances in the kidney regeneration including the manipulation of renal tubular cells, fibroblasts, endothelial cells, and macrophages in renal disease. Several types of stem cells, such as bone marrow-derived cells, adipocyte-derived mesenchymal stem cells, embryonic stem cells, and induced pluripotent stem cells are also applied for renal regeneration. Endogenous or lineage reprogrammed renal progenitor cells represent an attractive possibility for differentiation into multiple renal cell types. Angiogenesis can ameliorate hypoxia and renal fibrosis. Based on these studies and knowledge, we hope to innovate more reliable pharmacological or biotechnical methods for kidney regeneration medicine.
Collapse
|
14
|
Takaori K, Yanagita M. Kidney regeneration and stem cells. Anat Rec (Hoboken) 2013; 297:129-36. [PMID: 24293404 DOI: 10.1002/ar.22801] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 09/13/2013] [Indexed: 12/31/2022]
Abstract
The kidney has the capacity to recover from ischemic and toxic insults. Although there has been debate about the origin of cells that replace injured epithelial cells, it is now widely recognized that intrinsic surviving tubular cells are responsible for the repair. On the other hand, the cells, which have stem cell-like characteristics, have been isolated in the kidney using various methods, but it remains unknown if these stem cells actually exist in the adult kidney and if they are involved in kidney regeneration. This review will focus on the pathophysiology of kidney regeneration and the contribution of renal stem cells. We also discuss possible therapeutic applications to kidney disease.
Collapse
Affiliation(s)
- Koji Takaori
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | |
Collapse
|
15
|
Sun D, Bu L, Liu C, Yin Z, Zhou X, Li X, Xiao A. Therapeutic effects of human amniotic fluid-derived stem cells on renal interstitial fibrosis in a murine model of unilateral ureteral obstruction. PLoS One 2013; 8:e65042. [PMID: 23724119 PMCID: PMC3665750 DOI: 10.1371/journal.pone.0065042] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 04/25/2013] [Indexed: 11/18/2022] Open
Abstract
Interstitial fibrosis is regarded as the main pathway for the progression of chronic kidney disease (CKD) and is often associated with severe renal dysfunction. Stem cell-based therapies may provide alternative approaches for the treatment of CKD. Human amniotic fluid-derived stem cells (hAFSCs) are a novel stem cell population, which exhibit both embryonic and mesenchymal stem cell characteristics. Herein, the present study investigated whether the transplantation of hAFSCs into renal tissues could improve renal interstitial fibrosis in a murine model of unilateral ureteral obstruction (UUO). We showed that hAFSCs provided a protective effect and alleviated interstitial fibrosis as reflected by an increase in microvascular density; additionally, hAFSCs treatment beneficially modulated protein levels of vascular endothelial growth factor (VEGF), hypoxia inducible factor-1α (HIF-1α) and transforming growth factor-β1 (TGF-β1). Therefore, we hypothesize that hAFSCs could represent an alternative, readily available source of stem cells that can be applied for the treatment of renal interstitial fibrosis.
Collapse
Affiliation(s)
- Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical College, Xuzhou, PR China.
| | | | | | | | | | | | | |
Collapse
|
16
|
Pino CJ, Yevzlin AS, Tumlin J, Humes HD. Cell-based strategies for the treatment of kidney dysfunction: a review. Blood Purif 2012; 34:117-23. [PMID: 23095410 PMCID: PMC3836365 DOI: 10.1159/000341649] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Conventional treatment of acute and chronic renal diseases has focused on solute removal. Novel strategies aim to treat the multifactorial disease states of acute kidney injury and chronic kidney disease by mitigating inflammation. Cell-based technologies for the treatment of kidney dysfunction fall under two broad categories: cell therapy and cell processing. Cell therapy utilizes cells that are isolated, cultured outside of the body, and reintroduced as therapy, leveraging beneficial metabolic and synthetic functions. For example, renal tubule cells have been used to provide gluconeogenesis, ammoniagenesis, metabolism of glutathione, catabolism of important peptide hormones, growth factors, and cytokines critical to multiorgan homeostasis and immunomodulation to treat renal dysfunction. Cell processing focuses on altering the characteristics of cell populations inside the body to provide therapy. The selective cytopheretic device is an example of this novel therapeutic strategy that aims to modulate the innate immune response during organ dysfunction, additional organ injury, by binding and deactivating leukocytes. In this review, both cell therapy and cell processing approaches will be discussed in the context of acute kidney injury and chronic renal disease.
Collapse
|
17
|
Yokoo T, Fukui A, Kobayashi E. Application of regenerative medicine for kidney diseases. Organogenesis 2012; 3:34-43. [PMID: 19279698 DOI: 10.4161/org.3.1.3961] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Following recent advancements of stem cell research, the potential for organ regeneration using somatic stem cells as an ultimate therapy for organ failure has increased. However, anatomically complicated organs such as the kidney and liver have proven more refractory to stem cell-based regenerative techniques. At present, kidney regeneration is considered to require one of two approaches depending on the type of renal failure, namely acute renal failure (ARF) and chronic renal failure (CRF).The kidney has the potential to regenerate itself provided that the damage is not too severe and the kidney's structure remains intact. Regenerative medicine for ARF should therefore aim to activate or support this potent. In cases of the irreversible damage to the kidney, which is most likely in patients with CRF undergoing long-term dialysis, self-renewal is totally lost. Thus, regenerative medicine for CRF will likely involve the establishment of a functional whole kidney de novo. This article reviews the challenges and recent advances in both approaches and discusses the potential approach of these novel strategies for clinical application.
Collapse
Affiliation(s)
- Takashi Yokoo
- Division of Nephrology and Hypertension; Department of Internal Medicine; The Jikei University School of Medicine; Tokyo, Japan
| | | | | |
Collapse
|
18
|
Du T, Cheng J, Zhong L, Zhao XF, Zhu J, Zhu YJ, Liu GH. The alleviation of acute and chronic kidney injury by human Wharton's jelly-derived mesenchymal stromal cells triggered by ischemia-reperfusion injury via an endocrine mechanism. Cytotherapy 2012; 14:1215-27. [PMID: 22920838 DOI: 10.3109/14653249.2012.711471] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND AIMS The effects of human Wharton's jelly-derived mesenchymal stromal cells (WJ-MSC) on acute and chronic kidney injury induced by ischemia-reperfusion injury (IRI) were assessed. METHODS WJ-MSC were injected intravenously immediately after solitary kidney ischemia for 45 min. Cells were labeled with 5-bromo-2'deoxy-uridine (BrdU) for tracing in vivo. At 48 h post-IRI, serum creatinine and blood urea nitrogen (BUN) were measured. Tubular cell proliferation and apoptosis as well as activation of the Akt signal were identified by immunostaining. Real-time polymerase chain reaction (PCR) was employed to determine gene expression of inflammation-related cytokines and hepatocyte growth factor (HGF). Levels of human HGF were assayed by enzyme-linked immunosorbant assay (ELISA). Twenty-two weeks later, renal fibrosis was assessed by Masson's tri-chrome staining, collagen content and α-smooth muscle actin (α-SMA) staining. RESULTS There was no sign of labeled cells residing in the damaged kidney. Acute renal dysfunction elicited by IRI was considerably improved by WJ-MSC, in parallel with a stronger proliferative response and less apoptotic events. Additionally, phosphoAkt staining in injured tubular cells was substantially intensified. Cell treatment also caused a remarkable up-regulation of kidney interleukin (IL)-10, heme oxygenase (HO)-1 and HGF expression. Human HGF was detected in cell supernatants and the serum of cell-infused rats. Moreover, IRI-initiated fibrosis was abrogated by cell therapy, coincident with function amelioration. CONCLUSIONS WJ-MSC alleviate acute kidney injury, thereby rescuing the ensuing fibrotic lesions in an endocrine manner. The Akt signal in impaired tubular cells is reinforced by WJ-MSC, facilitating cell resistance to apoptosis and cell proliferation. HGF, either delivered or induced by WJ-MSC, is an important contributor.
Collapse
Affiliation(s)
- Tao Du
- Department of Urology, Shanghai Jiaotong University School of Medicine, Shanghai, PR China
| | | | | | | | | | | | | |
Collapse
|
19
|
Coussa RG, Shah S, Jain P, Martoni C, Bhathena J, Malhotra M, Prakash S. Microencapsulated Saccharomyces cerevisiae column bioreactor for potential use in renal failure uremia. ARTIFICIAL CELLS, BLOOD SUBSTITUTES, AND IMMOBILIZATION BIOTECHNOLOGY 2012; 40:103-12. [PMID: 22288841 DOI: 10.3109/10731199.2011.597758] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
A novel bioreactor containing viable APA microencapsulated yeast cells was designed. Rat plasma was used for perfusion. Yeast cell loading and perfusion flow rate were studied to maximize urea removal. An increase in column loading from 25% to 100%, increased urea removal from 5.67 ± 1.34% to 30.45 ± 0.48%. An increase in flow rate from low to high, increased urea removal from 30.46% to 40.4%. At 100% column loading and high flow rate, the creatinine and phosphate concentrations decreased by 22% and 10%, respectively, while ammonia concentrations increased by 58.9% (p < 0.05). Our in-vitro perfusion study demonstrates that microencapsulated yeast cells can remove urea efficiently.
Collapse
Affiliation(s)
- Razek Georges Coussa
- Department of Biomedical Engineering, Faculty of Medicine, McGill University, Montreal, Quebec, Canada
| | | | | | | | | | | | | |
Collapse
|
20
|
Erk in kidney diseases. JOURNAL OF SIGNAL TRANSDUCTION 2011; 2011:768512. [PMID: 21776388 PMCID: PMC3135240 DOI: 10.1155/2011/768512] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2010] [Accepted: 02/01/2011] [Indexed: 11/17/2022]
Abstract
Acute or chronic kidney injury results from various insults and pathological conditions, and is accompanied by activation of compensatory repair mechanisms. Both insults and repair mechanisms are initiated by circulating factors, whose cellular effects are mediated by activation selective signal transduction pathways. Two main signal transduction pathways are activated during these processes, the phosphatidylinositol 3' kinase (PI-3K)/mammalian target of rapamycin (mTOR) and the mitogen-activated protein kinase (MAPK) cascades. This review will focus on the latter, and more specifically on the role of extracellular signal-regulated kinase (ERK) cascade in kidney injury and repair.
Collapse
|
21
|
Timsit MO, Yuan X, Floerchinger B, Ge X, Tullius SG. Consequences of transplant quality on chronic allograft nephropathy. Kidney Int 2011:S54-8. [PMID: 21116319 DOI: 10.1038/ki.2010.424] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Using kidneys from expanded-criteria donors to alleviate organ shortage has raised concern on reduced transplant outcomes. In this paper, we review how critical donor-related factors such as donor age, brain death, and consequences of ischemia-reperfusion injury (IRI) determine graft quality and impact chronic allograft nephropathy. We propose that combinatorial effects of organ-intrinsic features associated with increasing age and unspecific injuries related to brain death and IRI will impact innate and adaptive immune responses. Future research will need to explore avenues to optimize donor management, organ preservation, adapted immunosuppressive strategies, as well as modifications of the allocation of suboptimal allografts.
Collapse
Affiliation(s)
- Marc-Olivier Timsit
- Division of Transplant Surgery and Transplant Surgery Research Laboratory, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | |
Collapse
|
22
|
Therapeutic implications of mesenchymal stem cells transfected with hepatocyte growth factor transplanted in rat kidney with unilateral ureteral obstruction. J Pediatr Surg 2011; 46:537-45. [PMID: 21376206 DOI: 10.1016/j.jpedsurg.2010.09.040] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2010] [Revised: 08/31/2010] [Accepted: 09/20/2010] [Indexed: 12/28/2022]
Abstract
PURPOSE The purpose of the study was to establish whether bone marrow mesenchymal stem cells (MSCs) transfected with hepatocyte growth factor (HGF) can migrate and localize in the rat's kidney with unilateral ureteral obstruction (UUO) and contribute to repair of renal fibrosis. METHODS We separated and cultured bone marrow-derived MSCs of male rats in vitro and transfected them with adenovirus-mediated HGF (Ad-HGF). The expression of HGF was measured with enzyme-linked immunosorbent assay. Sixty female rats were sham operated (n = 24) or subjected to left UUO: Ad-HGF-transfected MSCs, uninfected MSCs, or saline was injected into the rat's tail vein. Kidney tissue was collected at the end of the seventh or 14th day after operation. The distribution of Y chromosome in the kidney after Ad-HGF-transfected MSCs transplantation was determined by an in situ hybridization method. As the hallmark of myofibroblasts, α-smooth muscle actin (expression of which significantly increases in the presence of renal fibrosis) was detected by immunohistochemistry in all UUO rats' left kidney tissue. RESULTS Y chromosome-positive cells were found only in the obstructed kidney of the transplantation group. The positive cells were mainly distributed in the tubular cells. The average intensity of immunolabeling for α-smooth muscle actin in the transplanted group significantly decreased compared with sham-transplanted group (P < .05), and the expression in the rats injected with uninfected MSCs was higher than that in the rats with MSCs transfected with HGF (P < .05). CONCLUSIONS Mesenchymal stem cells transfected with HGF can migrate to the rat kidney with UUO and are mainly distributed in the region of renal tubular epithelial cells. The data indicate that MSCs transfected with HGF contribute to a reduction of renal fibrosis after ureteral obstruction and suggest that this may be exploited therapeutically.
Collapse
|
23
|
Wu W, Dnyanmote AV, Nigam SK. Remote communication through solute carriers and ATP binding cassette drug transporter pathways: an update on the remote sensing and signaling hypothesis. Mol Pharmacol 2011; 79:795-805. [PMID: 21325265 DOI: 10.1124/mol.110.070607] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Recent data from knockouts, human disease, and transport studies suggest that solute carrier (SLC) and ATP binding cassette (ABC) multispecific "drug" transporters maintain effective organ and body fluid concentrations of key nutrients, signaling molecules, and antioxidants. These processes involve transcellular movement of solutes across epithelial barriers and fluid compartments (e.g., blood, cerebrospinal fluid, urine, bile) via "matching" or homologous sets of SLC (e.g., SLC21, SLC22, SLC47) and ABC transporters. As described in the "Remote Sensing and Signaling Hypothesis" (Biochem Biophys Res Commun 323:429-436, 2004; Biochem Biophys Res Commun 351:872-876, 2006; J Biol Chem 282:23841-23853, 2007; Nat Clin Pract Nephrol 3:443-448, 2007; Mol Pharmacol 76:481-490, 2009), highly regulated transporter networks with overlapping substrate preferences are involved in sensing and signaling to maintain homeostasis in response to environmental changes (e.g., substrate imbalance and injury). They function in parallel with (and interact with) the endocrine and autonomic systems. Uric acid (urate), carnitine, prostaglandins, conjugated sex steroids, cGMP, odorants, and enterobiome metabolites are discussed here as examples. Xenobiotics hitchhike on endogenous carrier systems, sometimes leading to toxicity and side effects. By regulation of the expression and/or function of various remote organ multispecific transporters after injury, the overall transport capacity of the remote organ to handle endogenous toxins, metabolites, and signaling molecules may change, aiding in recovery. Moreover, these transporters may play a role in communication between organisms. The specific cellular components involved in sensing and altering transporter abundance or functionality depend upon the metabolite in question and probably involve different types of sensors as well as epigenetic regulation.
Collapse
Affiliation(s)
- Wei Wu
- Department of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | | | | |
Collapse
|
24
|
Eliopoulos N, Zhao J, Bouchentouf M, Forner K, Birman E, Yuan S, Boivin MN, Martineau D. Human marrow-derived mesenchymal stromal cells decrease cisplatin renotoxicity in vitro and in vivo and enhance survival of mice post-intraperitoneal injection. Am J Physiol Renal Physiol 2010; 299:F1288-98. [DOI: 10.1152/ajprenal.00671.2009] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Acute kidney injury (AKI) can occur from the toxic side-effects of chemotherapeutic agents such as cisplatin. Bone marrow-derived mesenchymal stromal cells (MSCs) have demonstrated wide therapeutic potential often due to beneficial factors they secrete. The goal of this investigation was to evaluate in vitro the effect of human MSCs (hMSCs) secretome on cisplatin-treated human kidney cells, and in vivo the consequence of hMSCs intraperitoneal (ip) implantation in mice with AKI. Our results revealed that hMSCs-conditioned media improved survival of HK-2 human proximal tubular cells exposed to cisplatin in vitro. This enhanced survival was linked to increased expression of phosphorylated Akt (Ser473) and was reduced by a VEGF-neutralizing antibody. In vivo testing of these hMSCs established that ip administration in NOD-SCID mice decreased cisplatin-induced kidney function impairment, as demonstrated by lower blood urea nitrogen levels and higher survival. In addition, blood phosphorous and amylase levels were also significantly decreased. Moreover, hMSCs reduced the plasma levels of several inflammatory cytokines/chemokines. Immunohistochemical examination of kidneys showed less apoptotic and more proliferating cells. Furthermore, PCR indicated the presence of hMSCs in mouse kidneys, which also showed enhanced expression of phosphorylated Akt. In conclusion, our study reveals that hMSCs can exert prosurvival effects on renal cells in vitro and in vivo, suggests a paracrine contribution for kidney protective abilities of hMSCs delivered ip, and supports their clinical potential in AKI.
Collapse
Affiliation(s)
- Nicoletta Eliopoulos
- Lady Davis Institute for Medical Research and
- Department of Surgery, Division of Surgical Research, McGill University, Montreal; and
| | - Jing Zhao
- Lady Davis Institute for Medical Research and
| | | | | | | | - Shala Yuan
- Lady Davis Institute for Medical Research and
| | | | - Daniel Martineau
- Department of Veterinary Medicine, Université de Montréal, St.-Hyacinthe, Quebec, Canada
| |
Collapse
|
25
|
Basile DP, Friedrich JL, Spahic J, Knipe N, Mang H, Leonard EC, Changizi-Ashtiyani S, Bacallao RL, Molitoris BA, Sutton TA. Impaired endothelial proliferation and mesenchymal transition contribute to vascular rarefaction following acute kidney injury. Am J Physiol Renal Physiol 2010; 300:F721-33. [PMID: 21123492 DOI: 10.1152/ajprenal.00546.2010] [Citation(s) in RCA: 222] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Acute kidney injury induces the loss of renal microvessels, but the fate of endothelial cells and the mechanism of potential vascular endothelial growth factor (VEGF)-mediated protection is unknown. Cumulative cell proliferation was analyzed in the kidney of Sprague-Dawley rats following ischemia-reperfusion (I/R) injury by repetitive administration of BrdU (twice daily) and colocalization in endothelial cells with CD31 or cablin. Proliferating endothelial cells were undetectable for up to 2 days following I/R and accounted for only ∼1% of BrdU-positive cells after 7 days. VEGF-121 preserved vascular loss following I/R but did not affect proliferation of endothelial, perivascular cells or tubular cells. Endothelial mesenchymal transition states were identified by localizing endothelial markers (CD31, cablin, or infused tomato lectin) with the fibroblast marker S100A4. Such structures were prominent within 6 h and sustained for at least 7 days following I/R. A Tie-2-cre transgenic crossed with a yellow fluorescent protein (YFP) reporter mouse was used to trace the fate of endothelial cells and demonstrated interstititial expansion of YFP-positive cells colocalizing with S100A4 and smooth muscle actin following I/R. The interstitial expansion of YFP cells was attenuated by VEGF-121. Multiphoton imaging of transgenic mice revealed the alteration of YFP-positive vascular cells associated with blood vessels characterized by limited perfusion in vivo. Taken together, these data indicate that vascular dropout post-AKI results from endothelial phenotypic transition combined with an impaired regenerative capacity, which may contribute to progressive chronic kidney disease.
Collapse
Affiliation(s)
- David P Basile
- Department of Cellular and Integrative Physiology, Indiana Center for Biological Microscopy, Indiana University, Indianapolis, Indiana.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Pino CJ, Humes HD. Stem cell technology for the treatment of acute and chronic renal failure. Transl Res 2010; 156:161-8. [PMID: 20801413 PMCID: PMC2930898 DOI: 10.1016/j.trsl.2010.07.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 07/14/2010] [Accepted: 07/15/2010] [Indexed: 11/16/2022]
Abstract
Acute and chronic renal failure are disorders with high rates of morbidity and mortality. Current treatment is based upon conventional dialysis to provide volume regulation and small solute clearance. There is growing recognition that renal failure is a complex disease state requiring a multifactorial therapy to address the short-comings of the conventional monofactorial approach. Kidney transplantation remains the most effective treatment, however, organ availability lags far behind demand. Many key kidney functions including gluconeogenesis, ammoniagenesis, metabolism of glutathione, catabolism of important peptide hormones, growth factors, and cytokines critical to multiorgan homeostasis and immunomodulation are provided by renal tubule cells. Therefore, cell-based therapies are promising multifactorial treatment approaches. In this review, current stem cell technologies including adult stem cells, embryonic stem cells and induced pluripotent stem cells will be discussed as cell sources for the treatment of acute and chronic renal failure.
Collapse
|
27
|
Oral microencapsulated live Saccharomyces cerevisiae cells for use in renal failure uremia: preparation and in vivo analysis. J Biomed Biotechnol 2010; 2010. [PMID: 20798777 PMCID: PMC2926829 DOI: 10.1155/2010/620827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 04/21/2010] [Indexed: 01/26/2023] Open
Abstract
Orally administrable alginate-poly-L-lysine-alginate (APA) microcapsules containing live yeast cells was investigated for use in renal failure. At all times, yeast cells remain inside the microcapsules, which are then excreted in the stool. During their gastrointestinal passage, small molecules, like urea, diffuse into the yeast microcapsules where they are hydrolyzed. Orally administrating these microcapsules to uremic rats was found to decrease urea concentrations from 7.29 ± 0.89 mmol/L to 6.12 ± 0.90 mmol/L over a treatment period of eight weeks. After stopping the treatment, the urea concentrations increased back to uremic levels of 7.64 ± 0.77 mmol/L. The analysis of creatinine concentrations averaged 39.19 ± 4.33 μmol/L, 50.83 ± 5.55 μmol/L, and 50.28 ± 7.10 μmol/L for the normal-control, uremic-control and uremic-treatment groups, respectively. While creatinine concentrations for both uremic-control and uremic-treatment groups did not differ among each other (P > .05), they were, however, significantly higher than those of the normal control group (P < .05). Uric acid concentrations averaged 80.08 ± 26.49 μmol/L, 99.92 ± 26.55 μmol/L, and 86.49 ± 28.42 μmol/L for the normal-control, uremic-control and uremic-treatment groups, respectively. There were no significant differences in both calcium and phosphate concentrations among all three groups (P > .05). The microbial populations of five tested types of bacteria were not substantially altered by the presence of the yeast APA encapsulated yeast (P > .05).
Collapse
|
28
|
Vicente-Vicente L, Quiros Y, Pérez-Barriocanal F, López-Novoa JM, López-Hernández FJ, Morales AI. Nephrotoxicity of uranium: pathophysiological, diagnostic and therapeutic perspectives. Toxicol Sci 2010; 118:324-47. [PMID: 20554698 DOI: 10.1093/toxsci/kfq178] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
As in the case of other heavy metals, a considerable body of evidence suggests that overexposure to uranium may cause pathological alterations to the kidneys in both humans and animals. In the present work, our aim was to analyze the available data from a critical perspective that should provide a view of the real danger of the nephrotoxicity of this metal for human beings. A further aim was to elaborate a comparative compilation of the renal pathophysiological data obtained in humans and experimental animals with a view to gaining more insight into our knowledge of the mechanisms of action and renal damage. Finally, we address the existing perspectives for the improvement of diagnostic methods and the treatment of intoxications by uranium, performing an integrated analysis of all these aspects.
Collapse
Affiliation(s)
- Laura Vicente-Vicente
- Unidad de Toxicología, Universidad de Salamanca, Edificio Departamental, S-19, Campus Miguel de Unamuno, 37007 Salamanca, Spain
| | | | | | | | | | | |
Collapse
|
29
|
Liao XH, Zhang L, Liu Q, Sun H, Peng CM, Guo H. Augmenter of liver regeneration protects kidneys from ischaemia/reperfusion injury in rats. Nephrol Dial Transplant 2010; 25:2921-9. [PMID: 20332418 DOI: 10.1093/ndt/gfq151] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Augmenter of liver regeneration (ALR), which was identified originally for its crucial role in promoting hepatocyte proliferation, is expressed in both the liver and kidney. Protective effects of ALR have been demonstrated in experimental models of acute liver failure. In the present study, we investigated the effect of ALR on renal ischaemia/reperfusion (I/R) injury and the possible mechanisms of its action. METHODS Male Sprague-Dawley rats were subjected to renal ischaemia for 60 min and then administered with either saline or recombinant human ALR (rhALR). A sham-operated group served as control. The expression of ALR in the sham-operated and acute kidney injury (AKI) groups was detected by immunohistochemistry and western blotting. Renal dysfunction and injury were assessed by measurement of serum biochemical markers and histological grading. Expression of proliferating cell nuclear antigen (PCNA) was determined by immunohistochemistry. RESULTS Renal ALR expression increased significantly in rats with ischaemic AKI compared with the sham-operated rats. Serum biochemical parameters showed that renal dysfunction was improved by administration of rhALR. Histological analysis revealed that treatment with rhALR also reduced the extent of kidney injury. Intraperitoneal injection of rhALR enhanced the proliferation of renal tubular cells. Conclusions. Administration of rhALR effectively reduces tubular injury and ameliorates the impairment of renal function. The protective effect of rhALR is associated with enhancement of renal tubular cell regeneration.
Collapse
Affiliation(s)
- Xiao-hui Liao
- Department of Nephrology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | | | | | | | | | | |
Collapse
|
30
|
Venkatachalam MA, Griffin KA, Lan R, Geng H, Saikumar P, Bidani AK. Acute kidney injury: a springboard for progression in chronic kidney disease. Am J Physiol Renal Physiol 2010; 298:F1078-94. [PMID: 20200097 DOI: 10.1152/ajprenal.00017.2010] [Citation(s) in RCA: 387] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Recently published epidemiological and outcome analysis studies have brought to our attention the important role played by acute kidney injury (AKI) in the progression of chronic kidney disease (CKD) to end-stage renal disease (ESRD). AKI accelerates progression in patients with CKD; conversely, CKD predisposes patients to AKI. This research gives credence to older, well-thought-out wisdom that recovery from AKI is often not complete and is marked by residual structural damage. It also mirrors older experimental observations showing that unilateral nephrectomy, a surrogate for loss of nephrons by disease, compromises structural recovery and worsens tubulointerstitial fibrosis after ischemic AKI. Moreover, review of a substantial body of work on the relationships among reduced renal mass, hypertension, and pathology associated with these conditions suggests that impaired myogenic autoregulation of blood flow in the setting of hypertension, the arteriolosclerosis that results, and associated recurrent ischemic AKI in microscopic foci play important roles in the development of progressively increasing tubulointerstitial fibrosis. How nutrition, an additional factor that profoundly affects renal disease progression, influences these events needs reevaluation in light of information on the effects of calories vs. protein and animal vs. vegetable protein on injury and progression. Considerations based on published and emerging data suggest that a pathology that develops in regenerating tubules after AKI characterized by failure of differentiation and persistently high signaling activity is the proximate cause that drives downstream events in the interstitium: inflammation, capillary rarefaction, and fibroblast proliferation. In light of this information, we advance a comprehensive hypothesis regarding the pathophysiology of AKI as it relates to the progression of kidney disease. We discuss the implications of this pathophysiology for developing efficient therapeutic strategies to delay progression and avert ESRD.
Collapse
Affiliation(s)
- Manjeri A Venkatachalam
- Dept. of Pathology, Univ. of Texas Health Science Center, 7703 Floyd Curl Dr., San Antonio, TX 78229, USA.
| | | | | | | | | | | |
Collapse
|
31
|
|
32
|
Lieberthal W, Levine JS. The role of the mammalian target of rapamycin (mTOR) in renal disease. J Am Soc Nephrol 2009; 20:2493-502. [PMID: 19875810 DOI: 10.1681/asn.2008111186] [Citation(s) in RCA: 228] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) is a serine/threonine kinase that plays a pivotal role in mediating cell size and mass, proliferation, and survival. mTOR has also emerged as an important modulator of several forms of renal disease. mTOR is activated after acute kidney injury and contributes to renal regeneration and repair. Inhibition of mTOR with rapamycin delays recovery of renal function after acute kidney injury. Activation of mTOR within the kidney also occurs in animal models of diabetic nephropathy and other causes of progressive kidney disease. Rapamycin ameliorates several key mechanisms believed to mediate changes associated with the progressive loss of GFR in chronic kidney disease. These include glomerular hypertrophy, intrarenal inflammation, and interstitial fibrosis. mTOR also plays an important role in mediating cyst formation and enlargement in autosomal dominant polycystic kidney disease. Inhibition of mTOR by rapamycin or one of its analogues represents a potentially novel treatment for autosomal dominant polycystic kidney disease. Finally, inhibitors of mTOR improve survival in patients with metastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Wilfred Lieberthal
- Stony Brook Medical Center, Health Sciences Center, 16-081B Nicholls Road, Stony Brook, NY 11794-8166, USA.
| | | |
Collapse
|
33
|
Watanabe N, Kato M, Suzuki N, Inoue C, Fedorova S, Hashimoto H, Maruyama S, Matsuo S, Wakamatsu Y. Kidney regeneration through nephron neogenesis in medaka. Dev Growth Differ 2009; 51:135-43. [PMID: 19207184 DOI: 10.1111/j.1440-169x.2009.01090.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Although renal regeneration is limited to repair of the proximal tubule in mammals, some bony fish are capable of renal regeneration through nephron neogenesis in the event of renal injury. We previously reported that nephron development in the medaka mesonephros is characterized by four histologically distinct stages, generally referred to as condensed mesenchyme, nephrogenic body, relatively small nephron, and the mature nephron. Developing nephrons are positive for wt1 expression during the first three of these stages. In the present study, we examined the regenerative response to renal injury, artificially induced by the administration of sublethal amounts of gentamicin in adult medaka. Similar to previous reports in other animals, the renal tubular epithelium and the glomerulus of the medaka kidney exhibited severe damage after exposure to this agent. However, kidneys showed substantial recovery after gentamicin administration, and a significant number of developing nephrons appeared 14 days after gentamicin administration (P < 0.01). Similarly, the expression of wt1 in developing nephrons also indicated the early stages of nephrogenesis. These findings show that medaka has the ability to regenerate kidney through nephron neogenesis during adulthood and that wt1 is a suitable marker for detecting nephrogenesis.
Collapse
Affiliation(s)
- Naoki Watanabe
- Bioscience and Biotechnology Center, Nagoya University, Chikusa-ku, Nagoya, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Liao XH, Zhang L, Tang XP, Liu Q, Sun H. Expression of augmenter of liver regeneration in rats with gentamicin-induced acute renal failure and its protective effect on kidney. Ren Fail 2009; 31:946-55. [PMID: 20030531 DOI: 10.3109/08860220903216154] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Augmenter of liver regeneration (ALR) enhances the proliferation of hepatocytes and accelerates recovery from acute liver failure in animal models. ALR is expressed in both the liver and kidney; however, the specific location of ALR expression and its biological effects in the kidney remain unknown. We aimed to investigate the efficacy of ALR in rats with gentamicin (GM)-induced acute renal failure (ARF). Rats were randomized into the normal group, GM+saline group, GM+vehicle group, and GM+rrALR group. Blood urea nitrogen, serum creatinine, and urine beta-N-acetyl-D-glucosaminidase were measured, and histological analyses of the kidneys were performed. The expression of ALR protein was determined by immunohistochemistry and Western blotting. In vitro incorporation of tritiated thymidine was used to measure the proliferation of renal tubular epithelial cells. In normal rats, the expression of ALR protein was faint in the medulla and absent in the cortex. However, in ARF rats, ALR expression increased significantly in both the renal cortex and medulla. Histological analyses revealed that treatment with recombinant rat ALR (rrALR) reduced the extent of injury of tubular cells in the renal cortex. Serum/urine biochemical parameters also showed that renal dysfunction was improved by the administration of rrALR. Intraperitoneal injection of rrALR enhanced the proliferation of tubular cells in vivo. We also confirmed that rrALR could promote the proliferation of renal tubular cells in vitro. These results indicate that rrALR effectively accelerates kidney recovery after ARF induced by gentamicin, and that the protective effect is associated with enhanced proliferation of renal tubular cells.
Collapse
Affiliation(s)
- Xiao-Hui Liao
- Department of Nephrology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | | | | | | | | |
Collapse
|
35
|
Xing J, Zhang Z, Mao H, Schnellmann RG, Zhuang S. Src regulates cell cycle protein expression and renal epithelial cell proliferation via PI3K/Akt signaling-dependent and -independent mechanisms. Am J Physiol Renal Physiol 2008; 295:F145-52. [PMID: 18434386 PMCID: PMC2494517 DOI: 10.1152/ajprenal.00092.2008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2008] [Accepted: 04/17/2008] [Indexed: 11/22/2022] Open
Abstract
Our recent studies showed that Src family kinases (SFKs) are important mediators of proliferation in renal proximal tubular cells (RPTC). In this study, we elucidate the signaling mechanisms that mediate SFK regulation of cell proliferation and cycle protein expression, and identify the SFK member responsible for these responses in a mouse RPTC line. Akt, a target of phosphoinositide-3-kinase (PI3K), and ERK1/2 were constitutively phosphorylated in RPTC cultured in the presence of serum. While treatment of cells with PP1, a specific SFK inhibitor, completely blocked phosphorylation of ERK1/2 and Akt, only inhibition of PI3K/Akt resulted in decreased RPTC proliferation. Incubation of cells with PP1 decreased cyclin D1 expression, decreased p27 and p57 phosphorylation, and increased p27 and p57 expression, two cyclin-dependent kinase inhibitors. Inhibition of the PI3K pathway decreased expression of cyclin D1 without altering expression of p27 and p57. In contrast, PP1 and PI3K inhibition had no effect on cyclin E and p21. Although RPTC expressed Src, Fyn, and Lyn, only siRNA-mediated knockdown of Src decreased RPTC proliferation, decreased cyclin D1 expression, and increased p27 and p57 expression. These data reveal that Src is a crucial mediator of RPTC proliferation and Src-mediated proliferation is associated with PI3K-dependent upregulation of cyclin D1 and PI3K-independent downregulation of p27 and p57.
Collapse
Affiliation(s)
- Jingping Xing
- Department of Medicine, Brown University School of Medicine, Rhode Island Hospital Middle, Providence, RI 02903, USA
| | | | | | | | | |
Collapse
|
36
|
Abstract
OBJECTIVE To review the cellular and molecular mechanisms of renal repair and recovery after acute kidney injury (AKI). DATA SOURCE The data were summarized from published research articles. RESULTS In AKI, there is an acute inflammatory response, epithelial cell necrosis and apoptosis, and shedding of epithelial cells into the tubular lumen. Recent work demonstrates that repopulation of damaged renal tubules occurs primarily from proliferation of tubular epithelial cells and resident renal-specific stem cells, with some contribution of paracrine factors from bone marrow-derived mesenchymal stem cells. In addition, growth factors seem to play a critical role in the repair process in animal models of renal injury. However, attempts to use growth factors in the clinical setting to attenuate human AKI or accelerate renal repair have not yet been successful. The endothelium also plays a critical role in the pathogenesis of AKI. Lastly, in human studies, the effect of dialysis on renal recovery remains poorly understood. CONCLUSIONS Experimental animal models of AKI demonstrate that renal recovery and repair involves proliferation of tubular epithelial cells and stem cell populations and the coordinated contribution of multiple growth factors. Future efforts to improve recovery from AKI and improve patient outcomes may include novel therapies based on manipulation of populations of stem cells and augmenting repopulation of renal tubules.
Collapse
|
37
|
Lash LH. Epidermal growth factor receptor ligands and renal epithelial cell proliferation. Am J Physiol Renal Physiol 2008; 294:F457-8. [DOI: 10.1152/ajprenal.00025.2008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
38
|
Zhuang S, Kinsey GR, Rasbach K, Schnellmann RG. Heparin-binding epidermal growth factor and Src family kinases in proliferation of renal epithelial cells. Am J Physiol Renal Physiol 2008; 294:F459-68. [PMID: 18171996 DOI: 10.1152/ajprenal.00473.2007] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Our recent studies have shown that proliferation of renal proximal tubular cells (RPTC) in the absence of growth factors requires activation of the epidermal growth factor (EGF) receptor. We sought to identify the endogenous EGF receptor ligand and investigate the mechanism(s) by which RPTC proliferate in different models. RPTC expressed both pro- and cleaved forms of heparin-binding epidermal growth factor (HB-EGF) and several metalloproteinases (MMP-2, -3, -9, and ADAM10, ADAM17) that have been reported to cleave HB-EGF. Treatment of RPTC with CRM 197, an inhibitor of HB-EGF binding to the EGF receptor, or downregulation of HB-EGF with small interfering RNA inhibited RPTC proliferation following plating. Furthermore, GM6001 (pan-MMP inhibitor), tumor-necrosis factor protease inhibitor-1 (TAPI-1; MMP and ADAM17 inhibitor), and GW280264X (ADAM10 and -17 inhibitor), but not GI254023X (ADAM10 inhibitor), attenuated the proliferation after plating. Although EGF receptor activation is required for RPTC proliferation after oxidant injury, CRM197, GM6001, and TAPI-1 did not block this response. In contrast, inhibition of Src with PP1 blocked EGF receptor activation and RPTC proliferation after oxidant injury. In addition, PP1 treatment attenuated HB-EGF-enhanced RPTC proliferation. We suggest that RPTC proliferation after plating is mediated by HB-EGF produced through an autocrine/paracrine mechanism and RPTC proliferation following oxidant injury is mediated by Src without involvement of HB-EGF.
Collapse
Affiliation(s)
- Shougang Zhuang
- Department of Medicine, Brown University School of Medicine, Rhode Island Hospital-Middle House 301, 593 Eddy St., Providence, RI 02903, USA.
| | | | | | | |
Collapse
|
39
|
Lechner J, Malloth NA, Jennings P, Heckl D, Pfaller W, Seppi T. Opposing roles of EGF in IFN-α-induced epithelial barrier destabilization and tissue repair. Am J Physiol Cell Physiol 2007; 293:C1843-50. [PMID: 17913840 DOI: 10.1152/ajpcell.00370.2007] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Balance between damaging influences and repair mechanisms determines the degree of tissue deterioration by inflammatory and other injury processes. Destabilization of the proximal tubular barrier has been previously shown to be induced by IFN-α, a cytokine crucial for linking innate and adaptive immune responses. EGF was implicated in rescue mechanisms from renal injury. To study the interplay between the two processes, we determined if EGF can prevent IFN-α-induced barrier permeabilization. EGF did not counteract but even exacerbated the IFN-α-induced decrease of transepithelial electrical resistance in LLC-PK1 monolayers. For this effect Erk1/2 activation was necessary, linking barrier regulation to EGF-induced cell cycle progression. In contrast to its damage-intensifying effect, EGF also facilitated the regeneration of epithelial barrier function after the termination of IFN-α treatment. This effect was not mediated by Erk1/2 activation or cell proliferation since U0126, an Erk1/2 inhibitor, did not prevent but ameliorated recovery. However, EGF accelerated the downregulation of caspase-3 in recovering cells. Similarly, a pan-caspase inhibitor was able to block caspase activity and, concomitantly, promote restoration of barrier function. Thus, barrier repair might be linked to an EGF-mediated antiapoptotic mechanism. EGF appears to sensitize epithelial cells to the detrimental effects of IFN-α but also helps to restore barrier function in the healing phase. The observed dual effect of EGF might be explained by the different impact of proproliferative and antiapoptotic signaling pathways during and after cytokine treatment. The timing of epithelial exposure to damaging agents and repair factors was identified as a crucial parameter determining tissue fate.
Collapse
Affiliation(s)
- Judith Lechner
- Division of Physiology, Department of Physiology and Medical Physics, Innsbruck Medical University, Fritz-Pregl-Strasse 3, Innsbruck, Austria.
| | | | | | | | | | | |
Collapse
|
40
|
Maeshima A. Label-retaining cells in the kidney: origin of regenerating cells after renal ischemia. Clin Exp Nephrol 2007; 11:269-274. [DOI: 10.1007/s10157-007-0500-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2007] [Accepted: 08/15/2007] [Indexed: 01/09/2023]
|
41
|
Ittrich H, Lange C, Tögel F, Zander AR, Dahnke H, Westenfelder C, Adam G, Nolte-Ernsting C. In vivo magnetic resonance imaging of iron oxide-labeled, arterially-injected mesenchymal stem cells in kidneys of rats with acute ischemic kidney injury: detection and monitoring at 3T. J Magn Reson Imaging 2007; 25:1179-91. [PMID: 17520738 DOI: 10.1002/jmri.20925] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE To evaluate MRI for a qualitative and quantitative in vivo tracking of intraaortal injected iron oxide-labeled mesenchymal stem cells (MSC) into rats with acute kidney injury (AKI). MATERIALS AND METHODS In vitro MRI and R2* measurement of nonlabeled and superparamagnetic iron oxide (SPIO)-labeled MSC (MSC(SPIO)) was performed in correlation to cellular iron content and cytological examination (Prussian blue, electron microscopy). In vivo MRI and R2* evaluation were performed before and after ischemic/reperfusion AKI (N = 14) and intraaortal injection of 1.5 x 10(6) MSC(SPIO) (N = 7), fetal calf serum (FCS) (medium, N = 6), and SPIO alone (N = 1) up to 14 days using a clinical 3T scanner. Signal to noise ratios (SNR), R2* of kidneys, liver, spleen, and bone marrow, renal function (creatinine [CREA], blood urea nitrogen [BUN]), and kidney volume were measured and tested for statistical significance (Student's t-test, P < 0.05) in comparison histology (hematoxylin and eosin [H&E], Prussian blue, periodic acid-Schiff [PAS], CD68). RESULTS In vitro, MSC(SPIO) showed a reduction of SNR and T2* with R2* approximately number of MSC(SPIO) (R2 = 0.98). In vivo MSC(SPIO) administration resulted in a SNR decrease (35 +/- 15%) and R2* increase (101 +/- 18.3%) in renal cortex caused by MSC(SPIO) accumulation in contrast to control animals (P < 0.01). Liver, spleen, and bone marrow (MSC(SPIO)) showed a delayed SNR decline/R2* increase (P < 0.05) resulting from MSC(SPIO) migration. The increase of kidney volume and the decrease in renal function (P < 0.05) was reduced in MSC-treated animals. CONCLUSION Qualitative and quantitative in vivo cell-tracking and monitoring of organ distribution of intraaortal injected MSC(SPIO) in AKI is feasible in MRI at 3T.
Collapse
Affiliation(s)
- Harald Ittrich
- Department of Diagnostic and Interventional Radiology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, DE-20246 Hamburg, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Zhuang S, Yan Y, Daubert RA, Schnellmann RG. Epiregulin promotes proliferation and migration of renal proximal tubular cells. Am J Physiol Renal Physiol 2007; 293:F219-26. [PMID: 17389679 DOI: 10.1152/ajprenal.00082.2007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Epiregulin is an epidermal growth factor (EGF) member that activates ErBB1 and ErBB4 homodimers and all possible heterodimeric ErbB complexes. Because its role in renal cell regeneration has not been investigated, we assessed the effect of exogenous epiregulin on regeneration of renal proximal tubular cells (RPTC) in primary culture. Epiregulin (10 ng/ml) was equivalent to EGF (10 ng/ml) in enhancing RPTC proliferation and migration. Epiregulin induced activation of the EGF receptor (EGFR), Akt, a downstream kinase of phosphoinositide 3-kinase (PI3K), and extracellular signaling-regulated kinase 1/2 (ERK1/2). Treatment with AG1478, a specific EGFR inhibitor, blocked phosphorylation of EGFR, Akt, ERK1/2, proliferation, and migration. Furthermore, inactivation of PI3K with LY-294002 blocked epiregulin-induced RPTC proliferation and, to a lesser extent, migration. However, blockade of ERK1/2 had no such effects. We suggest that epiregulin is a potent mitogen for renal epithelial cells and may contribute to renal regeneration through activation of EGFR and PI3/Akt pathways.
Collapse
Affiliation(s)
- Shougang Zhuang
- Dept. of Medicine, Brown University School of Medicine, Providence, RI 02903, USA.
| | | | | | | |
Collapse
|
43
|
Abstract
The kidney is a remarkable organ whose functions include maintaining fluid and electrolyte balance, excreting metabolic waste products, and controlling vascular tone. Blood flow within the kidney is very heterogeneous, which places the metabolically active medulla at high risk for ischemic injury. A number of mediators play a role in the modulation of renal blood flow, including angiotensin II, dopamine, vasopressin, prostaglandins, atrial natriuretic peptide, endothelin, and nitric oxide. Early markers of renal injury elicit strong interest, although currently there is no reliable marker available. Surgery causes the release of catecholamines, renin, angiotensin, and AVP that lead to a redistribution of renal blood flow and a decrease in GFR. Additionally, general anesthesia often results in some degree of hypotension and depressed cardiac output, which further reduces renal perfusion and potentially jeopardizes renal function. A careful anesthetic plan is imperative in the patient with renal insufficiency or failure because acute renal failure in the perioperative period is associated with a high morbidity and mortality. Factors including advanced age, diabetes, underlying renal insufficiency, and heart failure place a patient at high risk for developing acute renal failure. It is imperative to maintain euvolemia, normotension, and cardiac output, and to avoid nephrotoxic agents to optimize renal blood flow and renal perfusion as the best prevention of renal dysfunction. Further studies are needed to establish if any therapies exist to prevent or treat renal dysfunction effectively.
Collapse
Affiliation(s)
- Gebhard Wagener
- Department of Anesthesiology, College of Physicians and Surgeons at Columbia University, New York Presbyterian Hospital, Ph-5, 633 W. 168th Street, New York, NY 10032, USA
| | | |
Collapse
|
44
|
Litbarg NO, Gudehithlu KP, Sethupathi P, Arruda JAL, Dunea G, Singh AK. Activated omentum becomes rich in factors that promote healing and tissue regeneration. Cell Tissue Res 2007; 328:487-97. [PMID: 17468892 DOI: 10.1007/s00441-006-0356-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2006] [Accepted: 10/20/2006] [Indexed: 02/06/2023]
Abstract
In order to study the mechanism by which an omental pedicle promotes healing when applied to an injured site, we injected a foreign body into the abdominal cavity to activate the omentum. One week after the injection, we isolated the omentum and measured blood vessel density, blood content, growth and angiogenesis factors (VEGF and others), chemotactic factors (SDF-1 alpha), and progenitor cells (CXCR-4, WT-1). We found that the native omentum, which consisted mostly of adipose tissue, expanded the mass of its non-adipose part (milky spots) 15- to 20-fold. VEGF and other growth factors increased by two- to four-fold, blood vessel density by three-fold, and blood content by two-fold. The activated omentum also showed increases in SDF-1 alpha, CXCR-4, and WT-1 cells (factors and cells positively associated with tissue regeneration). Thus, we propose that an omentum activated by a foreign body (or by injury) greatly expands its milky-spot tissue and becomes rich in growth factors and progenitor cells that facilitate the healing and regeneration of injured tissue.
Collapse
Affiliation(s)
- Natalia O Litbarg
- Department of Medicine, Stroger Hospital of Cook County, Chicago, IL 60612, USA
| | | | | | | | | | | |
Collapse
|
45
|
Monte JC, Sakurai H, Bush KT, Nigam SK. The developmental nephrome: systems biology in the developing kidney. Curr Opin Nephrol Hypertens 2007; 16:3-9. [PMID: 17143064 DOI: 10.1097/mnh.0b013e3280118a5a] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW A set of important genes and signaling pathways involved in kidney development is emerging from analyses of mutant mice, in-vitro models, and global gene expression patterns. Conversion of data into dynamic models or networks through the synthesis of information at multiple levels is crucial for a better understanding of kidney development. RECENT FINDINGS Genetic and in-vitro evidence is beginning to provide a limited sense of the network topology in stages of kidney development. Intriguing data from other fields suggest how, with the aid of large-scale gene expression studies, these stages might be represented as dynamic attractor states. It is also suggested how branching morphogenesis of the epithelial ureteric bud may be sustained by an autocatalytic set of proteins whose interactions lead to repeated rounds of tip and stalk generation. Accumulating data in lower organisms suggest network topologies may be quite flexible, and the implications of these results for varieties of tubulogenesis and renal regeneration after acute injury are discussed. SUMMARY Currently it may be feasible to build tentative dynamic multistage models of nephrogenesis that facilitate experimental thinking. As data accumulate, it may become possible to test their predictive value.
Collapse
Affiliation(s)
- Julio C Monte
- Department of Medicine, Nephrology Division, Federal University of São Paulo, São Paulo, Brazil
| | | | | | | |
Collapse
|
46
|
Cheng CW, Rifai A, Ka SM, Shui HA, Lin YF, Lee WH, Chen A. Calcium-binding proteins annexin A2 and S100A6 are sensors of tubular injury and recovery in acute renal failure. Kidney Int 2006; 68:2694-703. [PMID: 16316344 DOI: 10.1111/j.1523-1755.2005.00740.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Rise in cellular calcium is associated with acute tubular necrosis, the most common cause of acute renal failure (ARF). The mechanisms that calcium signaling induce in the quiescent tubular cells to proliferate and differentiate during acute tubular necrosis have not been elucidated. METHODS Acute tubular necrosis induced in mice by single intravenous injection of uranyl nitrate and examined after 1, 3, 7, and 14 days. Renal function was monitored and kidneys were evaluated by histology, immunohistochemistry, Western blotting, in situ hybridization, and real-time reverse transcription-polymerase chain reaction (RT-PCR). Models of folic acid induced-ARF and ischemic/reperfusion (I/R) injury were similarly investigated. RESULTS Analysis of mRNA expression of intracellular calcium and phospholipid-binding proteins demonstrated selective expression of S100A6 and Annexin A2 (Anxa2) in the renal cortex with marked elevation on day 3, and gradually decline on day 7 and further attenuation on day 14. Similarly, the expression of both proteins, as demonstrated by immunohistochemistry and Western blot analysis, was increased and reached the peak level on day 7 and then gradually declined by day 14. Vimentin, a marker of dedifferentiated cells, was highly expressed during the recovery phase. Combined in situ hybridization immunohistochemistry revealed colocalization of both S100A6 and Anxa2 with proliferating cell nuclear antigen (PCNA). The universality of this phenomenon was confirmed in two other mouse acute tubular necrosis models, the ischemic-reperfusion injury and folic acid-induced ARF. CONCLUSION Collectively, these findings demonstrate that S100A6 and Anxa2 expression, initiated in response to tubular injury, persist in parallel throughout the recovery process of tubular cells in acute renal failure.
Collapse
Affiliation(s)
- Chao-Wen Cheng
- Graduate Institute of Life Sciences, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taiwan, Republic of China
| | | | | | | | | | | | | |
Collapse
|
47
|
Taulan M, Paquet F, Argiles A, Demaille J, Romey MC. Comprehensive analysis of the renal transcriptional response to acute uranyl nitrate exposure. BMC Genomics 2006; 7:2. [PMID: 16405725 PMCID: PMC1363347 DOI: 10.1186/1471-2164-7-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2005] [Accepted: 01/11/2006] [Indexed: 01/06/2023] Open
Abstract
Background Chemical and radiological toxicities related to uranium acute exposure have been widely studied in nuclear fuel workers and military personnel. It is well known that uranyl nitrate induces acute renal failure (ARF). However, the mechanisms of this metal-induced injury are not well defined at the molecular level. Results Renal function and histology were assessed in mice receiving uranyl nitrate (UN(+)) and controls (UN(-)). To identify the genomic response to uranium exposure, serial analysis gene expression (SAGE) of the kidney was performed in both groups. Over 43,000 mRNA SAGE tags were sequenced. A selection of the differentially expressed transcripts was confirmed by real-time quantitative PCR and Western blotting. UN(+) animals developed renal failure and displayed the characteristic histological lesions of UN nephropathy. Of the >14,500 unique tags identified in both libraries, 224 had a modified expression level; they are known to participate in inflammation, ion transport, signal transduction, oxidative stress, apoptosis, metabolism, and catabolism. Several genes that were identified had not previously been evaluated within the context of toxic ARF such as translationally controlled tumor protein, insulin like growth factor binding protein 7 and ribosomal protein S29, all apoptosis related genes. Conclusion We report a comprehensive description of the UN induced modifications in gene expression levels, including the identification of genes previously unrelated to ARF. The study of these genes and the metabolisms they control should improve our understanding of toxic ARF and enlighten on the molecular targets for potential therapeutic interventions.
Collapse
Affiliation(s)
- Magali Taulan
- Laboratoire de radiotoxicologie expérimentale, Institut de Radioprotection et de Sûreté Nucléaire, Site du Tricastin, BP 166 26702 Pierrelatte Cedex, France
- Laboratoire de Génétique Moléculaire, UPR 1142, Institut de Génétique Humaine, 141 Route de la Cardonille, 34396 Montpellier Cedex 05, France
| | - Francois Paquet
- Laboratoire de radiotoxicologie expérimentale, Institut de Radioprotection et de Sûreté Nucléaire, Site du Tricastin, BP 166 26702 Pierrelatte Cedex, France
| | - Angel Argiles
- Laboratoire de Génomique Fonctionnelle, UPR 2580, Institut de Génétique Humaine, 141 rue de la Cardonille, 34396 Montpellier Cedex 5, France
| | - Jacques Demaille
- Laboratoire de Génétique Moléculaire, UPR 1142, Institut de Génétique Humaine, 141 Route de la Cardonille, 34396 Montpellier Cedex 05, France
| | | |
Collapse
|
48
|
Maeshima A, Sakurai H, Nigam SK. Adult kidney tubular cell population showing phenotypic plasticity, tubulogenic capacity, and integration capability into developing kidney. J Am Soc Nephrol 2005; 17:188-98. [PMID: 16338966 DOI: 10.1681/asn.2005040370] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Using in vivo bromodeoxyuridine (BrdU) labeling, a tubular cell population (label-retaining tubular cells [LRTC]) was identified recently in normal adult kidneys, which contributes actively to the regeneration process of the kidney after injury. Here, these LRTC are characterized in vitro. The LRTC population was isolated from BrdU-treated rat kidney by FACS. Both LRTC and non-LRTC underwent proliferation and maintained an epithelial phenotype in the presence of tubulogenic growth factors such as EGF, TGF-alpha, IGF-I, and hepatocyte growth factor. It is interesting that LRTC also proliferated without epithelial markers expression in the presence of soluble factors derived from an embryonic kidney metanephric mesenchyme cell line. The type of extracellular matrix strongly influenced the phenotype of LRTC. Furthermore, in three-dimensional collagen gel culture, LRTC formed tubule-like or tubulocystic structures in response to growth factors (hepatocyte growth factor and fibroblast growth factor) that are known to induce kidney cell tubulogenesis in vitro and/or participate in renal regeneration in vivo. In contrast, non-LRTC did not form these structures. When transplanted into the metanephric kidney, LRTC but not non-LRTC were integrated into epithelial components of nephron, including the proximal tubular cells and the ureteric bud. They also differentiated into fibroblast-like cells. Collectively, these findings suggest that LRTC are an adult kidney tubular cell population that shows phenotypic plasticity, tubulogenic capacity, and integration capability into the developing kidney.
Collapse
Affiliation(s)
- Akito Maeshima
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0693, USA
| | | | | |
Collapse
|
49
|
Biju MP, Akai Y, Shrimanker N, Haase VH. Protection of HIF-1-deficient primary renal tubular epithelial cells from hypoxia-induced cell death is glucose dependent. Am J Physiol Renal Physiol 2005; 289:F1217-26. [PMID: 16048903 DOI: 10.1152/ajprenal.00233.2005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ischemic acute renal failure is a frequent clinical problem in hospitalized patients and is associated with significant mortality. Hypoxia-inducible factor 1 (HIF-1) mediates cellular adaptation to hypoxia by regulating biological processes important for cell survival, which include glycolysis, angiogenesis, erythropoiesis, apoptosis, and proliferation. To investigate the role of HIF-1 in hypoxia-induced renal epithelial cell death, we generated mice that allow inactivation of HIF-1α by tetracycline-inducible Cre-loxP-mediated recombination in primary renal proximal tubule cells (PRPTC), resulting in a suppression of HIF-1-mediated gene transcription during oxygen deprivation. In the absence of glucose, the onset and the degree of hypoxia-induced cell death in HIF-1-deficient PRPTC were comparable to wild-type cells. However, when glucose availability was limited, the onset of cell death was delayed in either PRPTC that were HIF-1 deficient or in wild-type PRPTC when glycolysis or glucose uptake was partially inhibited. Our findings suggest in an in vitro genetic model that 1) the generation of adequate energy levels for the maintenance of PRPTC viability under hypoxia does not require HIF-1 and 2) that HIF-1 regulates the timing of hypoxia-induced cell death and apoptosis onset through its effects on glucose consumption.
Collapse
Affiliation(s)
- Mangatt P Biju
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, 19104-6144, USA
| | | | | | | |
Collapse
|
50
|
Abstract
The inadequacy of current treatment modalities and insufficiency of donor organs for cadaveric transplantation have driven a search for improved methods of dealing with renal failure. The rising concept of cell-based therapeutics has provided a framework around which new approaches are being generated, and its combination with advances in stem cell research stands to bring both fields to clinical fruition. This budding partnership is presently in its very early stages, but an examination of the cell-based therapies currently under development clearly shows the magnitude of the role that stem cells will ultimately play. The issue over reports of unexpected plasticity in adult stem cell differentiation remains a focus of debate, and evidence for bone marrow-derived stem cell contributions to renal repair has been challenged. The search for adult renal stem cells, which could have a considerable impact on much of the work discussed here, appears to be narrowing. The use of embryonic tissue in research continues to provide valuable insights but will be the subject of intense societal scrutiny and debate before it reaches the stage of clinical application. Embryonic stem (ES) cells, with their ability to generate all, or nearly all, of the cell types in the adult body and a possible source of cells genetically identical to the donor, hold great promise but face ethical and political hurdles for human use. Immunoisolation of heterologous cells by encapsulation creates opportunities for their safe use as a component of implanted or ex vivo devices.
Collapse
Affiliation(s)
- James C Brodie
- Division of Nephrology, Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA.
| | | |
Collapse
|