1
|
Tavakolidakhrabadi N, Ding WY, Saleem MA, Welsh GI, May C. Gene therapy and kidney diseases. Mol Ther Methods Clin Dev 2024; 32:101333. [PMID: 39434922 PMCID: PMC11492605 DOI: 10.1016/j.omtm.2024.101333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Chronic kidney disease (CKD) poses a significant global health challenge, projected to become one of the leading causes of death by 2040. Current treatments primarily manage complications and slow progression, highlighting the urgent need for personalized therapies targeting the disease-causing genes. Our increased understanding of the underlying genomic changes that lead to kidney diseases coupled with recent successful gene therapies targeting specific kidney cells have turned gene therapy and genome editing into a promising therapeutic approach for treating kidney disease. This review paper reflects on different delivery routes and systems that can be exploited to target specific kidney cells and the ways that gene therapy can be used to improve kidney health.
Collapse
Affiliation(s)
- Nadia Tavakolidakhrabadi
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | - Wen Y. Ding
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | - Moin A. Saleem
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
- Department of Paediatric Nephrology, Bristol Royal Hospital for Children, University Hospitals Bristol and Weston NHS Foundation Trust, Upper Maudlin Street, Bristol BS2 8BJ, UK
| | - Gavin I. Welsh
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| | - Carl May
- Bristol Renal, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol BS1 3NY, UK
| |
Collapse
|
2
|
Henedak NT, El-Abhar HS, Soubh AA, Abdallah DM. NLRP3 Inflammasome: A central player in renal pathologies and nephropathy. Life Sci 2024; 351:122813. [PMID: 38857655 DOI: 10.1016/j.lfs.2024.122813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 05/16/2024] [Accepted: 06/04/2024] [Indexed: 06/12/2024]
Abstract
The cytoplasmic oligomer NLR Family Pyrin Domain Containing 3 (NLRP3) inflammasome has been implicated in most inflammatory and autoimmune diseases. Here, we highlight the significance of NLRP3 in diverse renal disorders, demonstrating its activation in macrophages and non-immune tubular epithelial and mesangial cells in response to various stimuli. This activation leads to the release of pro-inflammatory cytokines, contributing to the development of acute kidney injury (AKI), chronic renal injury, or fibrosis. In AKI, NLRP3 inflammasome activation and pyroptotic renal tubular cell death is driven by contrast and chemotherapeutic agents, sepsis, and rhabdomyolysis. Nevertheless, inflammasome is provoked in disorders such as crystal and diabetic nephropathy, obesity-related renal fibrosis, lupus nephritis, and hypertension-induced renal damage that induce chronic kidney injury and/or fibrosis. The mechanisms by which the inflammatory NLRP3/ Apoptosis-associated Speck-like protein containing a Caspase recruitment domain (ASC)/caspase-1/interleukin (IL)-1β & IL-18 pathway can turn on renal fibrosis is also comprehended. This review further outlines the involvement of dopamine and its associated G protein-coupled receptors (GPCRs), including D1-like (D1, D5) and D2-like (D2-D4) subtypes, in regulating this inflammation-linked renal dysfunction pathway. Hence, we identify D-related receptors as promising targets for renal disease management by inhibiting the functionality of the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Nada T Henedak
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, 6(th) of October City, Giza, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology, Toxicology, and Biochemistry, Faculty of Pharmacy, Future University in Egypt, Cairo 11835, Egypt
| | - Ayman A Soubh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ahram Canadian University, 6(th) of October City, Giza, Egypt
| | - Dalaal M Abdallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| |
Collapse
|
3
|
Cantero-Navarro E, Rayego-Mateos S, Orejudo M, Tejedor-Santamaria L, Tejera-Muñoz A, Sanz AB, Marquez-Exposito L, Marchant V, Santos-Sanchez L, Egido J, Ortiz A, Bellon T, Rodrigues-Diez RR, Ruiz-Ortega M. Role of Macrophages and Related Cytokines in Kidney Disease. Front Med (Lausanne) 2021; 8:688060. [PMID: 34307414 PMCID: PMC8295566 DOI: 10.3389/fmed.2021.688060] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/11/2021] [Indexed: 12/14/2022] Open
Abstract
Inflammation is a key characteristic of kidney disease, but this immune response is two-faced. In the acute phase of kidney injury, there is an activation of the immune cells to fight against the insult, contributing to kidney repair and regeneration. However, in chronic kidney diseases (CKD), immune cells that infiltrate the kidney play a deleterious role, actively participating in disease progression, and contributing to nephron loss and fibrosis. Importantly, CKD is a chronic inflammatory disease. In early CKD stages, patients present sub-clinical inflammation, activation of immune circulating cells and therefore, anti-inflammatory strategies have been proposed as a common therapeutic target for renal diseases. Recent studies have highlighted the plasticity of immune cells and the complexity of their functions. Among immune cells, monocytes/macrophages play an important role in all steps of kidney injury. However, the phenotype characterization between human and mice immune cells showed different markers; therefore the extrapolation of experimental studies in mice could not reflect human renal diseases. Here we will review the current information about the characteristics of different macrophage phenotypes, mainly focused on macrophage-related cytokines, with special attention to the chemokine CCL18, and its murine functional homolog CCL8, and the macrophage marker CD163, and their role in kidney pathology.
Collapse
Affiliation(s)
- Elena Cantero-Navarro
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Rayego-Mateos
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Macarena Orejudo
- Renal, Vascular and Diabetes Research Laboratory, Fundación IIS -Fundación Jiménez Díaz, Universidad Autónoma, Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Lucía Tejedor-Santamaria
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Tejera-Muñoz
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Belén Sanz
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
- Laboratory of Nephrology and Hypertension, Fundación IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Laura Marquez-Exposito
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Vanessa Marchant
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Laura Santos-Sanchez
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, Fundación IIS -Fundación Jiménez Díaz, Universidad Autónoma, Madrid, Spain
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Alberto Ortiz
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
- Laboratory of Nephrology and Hypertension, Fundación IIS-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
| | - Teresa Bellon
- La Paz Hospital Health Research Institute, Madrid, Spain
| | - Raúl R Rodrigues-Diez
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Ruiz-Ortega
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, Madrid, Spain
- Red de Investigación Renal, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
4
|
Song SJ, Kim SM, Lee SH, Moon JY, Hwang HS, Kim JS, Park SH, Jeong KH, Kim YG. Rhabdomyolysis-Induced AKI Was Ameliorated in NLRP3 KO Mice via Alleviation of Mitochondrial Lipid Peroxidation in Renal Tubular Cells. Int J Mol Sci 2020; 21:ijms21228564. [PMID: 33202867 PMCID: PMC7696646 DOI: 10.3390/ijms21228564] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022] Open
Abstract
Introduction: A recent study showed that early renal tubular injury is ameliorated in Nod-like receptor pyrin domain-containing protein 3 (NLRP3) KO mice with rhabdomyolysis-induced acute kidney injury (RIAKI). However, the precise mechanism has not been determined. Therefore, we investigated the role of NLRP3 in renal tubular cells in RIAKI. Methods: Glycerol-mediated RIAKI was induced in NLRP3 KO and wild-type (WT) mice. The mice were euthanized 24 h after glycerol injection, and both kidneys and plasma were collected. HKC-8 cells were treated with ferrous myoglobin to mimic a rhabdomyolytic environment. Results: Glycerol injection led to increase serum creatinine, aspartate aminotransferase (AST), and renal kidney injury molecule-1 (KIM-1) level; renal tubular necrosis; and apoptosis. Renal injury was attenuated in NLRP3 KO mice, while muscle damage and renal neutrophil recruitment did not differ between NLRP3 KO mice and WT mice. Following glycerin injection, increases in cleaved caspase-3, poly (ADP-ribose) polymerase (PARP), and a decrease in the glutathione peroxidase 4 (GPX-4) level were observed in the kidneys of mice with RIAKI, and these changes were alleviated in the kidneys of NLRP3 KO mice. NLRP3 was upregulated, and cell viability was suppressed in HKC-8 cells treated with ferrous myoglobin. Myoglobin-induced apoptosis and lipid peroxidation were significantly decreased in siNLRP3-treated HKC-8 cells compared to ferrous myoglobin-treated HKC-8 cells. Myoglobin reduced the mitochondrial membrane potential and increased mitochondrial fission and reactive oxygen species (ROS) and lipid peroxidation levels, which were restored to normal levels in NLRP3-depleted HKC-8 cells. Conclusions: NLRP3 depletion ameliorated renal tubular injury in a murine glycerol-induced acute kidney injury (AKI) model. A lack of NLRP3 improved tubular cell viability via attenuation of myoglobin-induced mitochondrial injury and lipid peroxidation, which might be the critical factor in protecting the kidney.
Collapse
|
5
|
Kim YG, Kim SM, Kim KP, Lee SH, Moon JY. The Role of Inflammasome-Dependent and Inflammasome-Independent NLRP3 in the Kidney. Cells 2019; 8:cells8111389. [PMID: 31694192 PMCID: PMC6912448 DOI: 10.3390/cells8111389] [Citation(s) in RCA: 107] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/23/2019] [Accepted: 10/30/2019] [Indexed: 01/27/2023] Open
Abstract
Cytoplasmic nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) forms an inflammasome with apoptosis-associated speck-like protein containing a CARD (ASC) and pro-caspase-1, which is followed by the cleavage of pro-caspase-1 to active caspase-1 and ultimately the activation of IL-1β and IL-18 and induction of pyroptosis in immune cells. NLRP3 activation in kidney diseases aggravates inflammation and subsequent fibrosis, and this effect is abrogated by genetic or pharmacologic deletion of NLRP3. Inflammasome-dependent NLRP3 mediates the progression of kidney diseases by escalating the inflammatory response in immune cells and the cross-talk between immune cells and renal nonimmune cells. However, recent studies have suggested that NLRP3 has several inflammasome-independent functions in the kidney. Inflammasome-independent NLRP3 regulates apoptosis in tubular epithelial cells by interacting with mitochondria and mediating mitochondrial reactive oxygen species production and mitophagy. This review will summarize the mechanisms by which NLRP3 functions in the kidney in both inflammasome-dependent and inflammasome-independent ways and the role of NLRP3 and NLRP3 inhibitors in kidney diseases.
Collapse
Affiliation(s)
- Yang Gyun Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Medical School, Seoul 02447, Korea; (Y.G.K.); (S.-M.K.); (S.-H.L.)
| | - Su-Mi Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Medical School, Seoul 02447, Korea; (Y.G.K.); (S.-M.K.); (S.-H.L.)
| | - Ki-Pyo Kim
- Division of Nephrology and Hypertension, Department of Internal Medicine, Inha University of Medicine, Incheon 22212, Korea;
| | - Sang-Ho Lee
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Medical School, Seoul 02447, Korea; (Y.G.K.); (S.-M.K.); (S.-H.L.)
| | - Ju-Young Moon
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University Medical School, Seoul 02447, Korea; (Y.G.K.); (S.-M.K.); (S.-H.L.)
- Correspondence: ; Tel.: +82-2-440-6262
| |
Collapse
|
6
|
Miao NJ, Xie HY, Xu D, Yin JY, Wang YZ, Wang B, Yin F, Zhou ZL, Cheng Q, Chen PP, Zhou L, Xue H, Zhang W, Wang XX, Liu J, Lu LM. Caspase-11 promotes renal fibrosis by stimulating IL-1β maturation via activating caspase-1. Acta Pharmacol Sin 2019; 40:790-800. [PMID: 30382182 DOI: 10.1038/s41401-018-0177-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 09/24/2018] [Indexed: 12/28/2022]
Abstract
Caspase-11 is a key upstream modulator for activation of inflammatory response under pathological conditions. In this study, we investigated the roles of caspase-11 in the maturation of interleukin-1β (IL-1β) and development of renal interstitial fibrosis in vivo and in vitro. Mice were subjected to unilateral ureteral obstruction (UUO). The mice were treated with either caspase-11 inhibitor wedelolactone (Wed, 30 mg/kg/day, ig) for 7 days or caspase-11 siRNA (10 nmol/20 g body weight per day, iv) for 14 days. The mice were euthanized on day 14, their renal tissue and blood sample were collected. We found that the obstructed kidney had significantly higher caspase-11 levels and obvious tubular injury and interstitial fibrosis. Treatment with Wed or caspase-11 siRNA significantly mitigated renal fibrosis in UUO mice, evidenced by the improved histological changes. Furthermore, caspase-11 inhibition significantly blunted caspase-1 activation, IL-1β maturation, transforming growth factor-β (TGF-β), fibronectin, and collagen I expressions in the obstructed kidney. Renal tubular epithelial NRK-52E cells were treated in vitro with angiotensin (Ang, 1 μmol/L), which stimulated caspase-11 activation and IL-1β maturation. Treatment with IL-1β (20 ng/ml) significantly increased the expression of TGF-β, fibronectin, and collagen I in the cells. Ang II-induced expression of TGF-β, fibronectin, and collagen I were suppressed by caspase-11 siRNA or Wed. Finally, we revealed using co-immunoprecipitation that caspase-11 was able to interact with caspase-1 in NRK-52E cells. These results suggest that caspase-11 is involved in UUO-induced renal fibrosis. Elevation of caspase-11 in the obstructed kidney promotes renal fibrosis by stimulating caspase-1 activation and IL-1β maturation.
Collapse
|
7
|
Mulay SR. Multifactorial functions of the inflammasome component NLRP3 in pathogenesis of chronic kidney diseases. Kidney Int 2019; 96:58-66. [PMID: 30922667 DOI: 10.1016/j.kint.2019.01.014] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 01/01/2019] [Accepted: 01/04/2019] [Indexed: 12/26/2022]
Abstract
The NLRP3 inflammasome is a cytosolic multiprotein caspase-activating complex platform involved in innate immunity required for the maturation and release of interleukin (IL)-1β and IL-18. Both cytokines activate their respective receptors present on cells inside and outside kidneys, resulting in the release of other proinflammatory cytokines to set up an inflammatory milieu both within the kidney and systemically. The canonical NLRP3-ASC-caspase-1-IL-1β-IL-18 axis has been shown to contribute to the pathophysiology of several kidney diseases by regulating renal necroinflammation. However, many recent studies have emphasized the inflammasome-independent functions of NLRP3 in chronic kidney disease (CKD) pathogenesis. This review highlights the contribution of the inflammasome-independent functions of NLPR3, for example, in fibrotic tissue remodeling, in tubular epithelial cell apoptosis, and in metabolic pathways, during the development and progression of CKD in various experimental models and humans. Interestingly, therapies targeting the inflammasome effectors (e.g., IL-1 receptor antagonists and IL-1β) have been approved for therapeutic use for NLRP3-dependent diseases; however, no NLRP3 antagonists have been approved for therapeutic use until now. This review highlights the double-edged sword-like functions of NLRP3 in the regulation of renal necroinflammation and fibrosis and therefore emphasizes the urgent need for specific NLRP3 inhibitors because of the broad therapeutic potential they offer for the treatment of CKD.
Collapse
Affiliation(s)
- Shrikant R Mulay
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow, India.
| |
Collapse
|
8
|
Abstract
The number of individuals affected by acute kidney injury (AKI) and chronic kidney disease (CKD) is constantly rising. In light of the limited availability of treatment options and their relative inefficacy, cell based therapeutic modalities have been studied. However, not many efforts are put into safety evaluation of such applications. The aim of this study was to review the existing published literature on adverse events reported in studies with genetically modified cells for treatment of kidney disease. A systematic review was conducted by searching PubMed and EMBASE for relevant articles published until June 2018. The search results were screened and relevant articles selected using pre-defined criteria, by two researchers independently. After initial screening of 6894 abstracts, a total number of 97 preclinical studies was finally included for full assessment. Of these, 61 (63%) presented an inappropriate study design for the evaluation of safety parameters. Only 4 studies (4%) had the optimal study design, while 32 (33%) showed sub-optimal study design with either direct or indirect evidence of adverse events. The high heterogeneity of studies included regarding cell type and number, genetic modification, administration route, and kidney disease model applied, combined with the consistent lack of appropriate control groups, makes a reliable safety evaluation of kidney cell-based therapies impossible. Only a limited number of relevant studies included looked into essential safety-related outcomes, such as inflammatory (48%), tumorigenic and teratogenic potential (12%), cell biodistribution (82%), microbiological safety with respect to microorganism contamination and latent viruses' reactivation (1%), as well as overall well-being and animal survival (19%). In conclusion, for benign cell-based therapies, well-designed pre-clinical studies, including all control groups required and good manufacturing processes securing safety, need to be done early in development. Preferably, this should be performed side by side with efficacy evaluation and according to the official guidelines of leading health organizations.
Collapse
|
9
|
Mylonas KJ, Anderson J, Sheldrake TA, Hesketh EE, Richards JA, Ferenbach DA, Kluth DC, Savill J, Hughes J. Granulocyte macrophage-colony stimulating factor: A key modulator of renal mononuclear phagocyte plasticity. Immunobiology 2018; 224:60-74. [PMID: 30415915 PMCID: PMC6401212 DOI: 10.1016/j.imbio.2018.10.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 10/26/2018] [Accepted: 10/30/2018] [Indexed: 02/07/2023]
Abstract
Macrophage-colony stimulating factor (M-CSF) and granulocyte macrophage-colony stimulating factor (GM-CSF) play key roles in the differentiation of macrophages and dendritic cells (DCs). We examined the effect of treatment with M-CSF-containing macrophage medium or GM-CSF-containing DC medium upon the phenotype of murine bone marrow-derived macrophages and DCs. Culture of macrophages for 5 days in DC medium reduced F4/80 expression and increased CD11c expression with cells effectively stimulating T cell proliferation in a mixed lymphocyte reaction. DC medium treatment of macrophages significantly reduced phagocytosis of both apoptotic cells and latex beads and strongly induced the expression of the chemokine receptor CCR7 known to be involved in DC trafficking to lymph nodes. Lysates of obstructed murine kidneys expressed both M-CSF and GM-CSF though M-CSF expression was dominant (M-CSF:GM-CSF ratio ∼30:1). However, combination treatment with both M-CSF and GM-CSF (ratio 30:1) indicated that small amounts of GM-CSF skewed macrophages towards a DC-like phenotype. To determine whether macrophage phenotype might be modulated in vivo we tracked CD45.1+ bone marrow-derived macrophages intravenously administered to CD45.2+ mice with unilateral ureteric obstruction. Flow cytometry of enzyme dissociated kidneys harvested 3 days later indicated CD11c and MHC Class II upregulation by adoptively transferred CD45.1+ cells with CD45.1+ cells evident in draining renal lymph nodes. Our data suggests that GM-CSF modulates mononuclear phagocyte plasticity, which likely promotes resolution of injury and healing in the injured kidney.
Collapse
Affiliation(s)
- Katie J Mylonas
- The University of Edinburgh/ Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent Edinburgh EH16 4TJ, Scotland, United Kingdom.
| | - Jennifer Anderson
- The University of Edinburgh/ Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Tara A Sheldrake
- The University of Edinburgh/ Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Emily E Hesketh
- The University of Edinburgh/ Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - James A Richards
- The University of Edinburgh/ Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - David A Ferenbach
- The University of Edinburgh/ Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - David C Kluth
- The University of Edinburgh/ Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - John Savill
- The University of Edinburgh/ Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent Edinburgh EH16 4TJ, Scotland, United Kingdom
| | - Jeremy Hughes
- The University of Edinburgh/ Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh BioQuarter, 47 Little France Crescent Edinburgh EH16 4TJ, Scotland, United Kingdom
| |
Collapse
|
10
|
Abstract
Kidney injury implies danger signaling and a response by the immune system. The inflammasome is a central danger recognition platform that triggers local and systemic inflammation. In immune cells, inflammasome activation causes the release of mature IL-1β and of the alarmin IL-1α Dying cells release IL-1α also, independently of the inflammasome. Both IL-1α and IL-1β ligate the same IL-1 receptor (IL-1R) that is present on nearly all cells inside and outside the kidney, further amplifying cytokine and chemokine release. Thus, the inflammasome-IL-1α/IL-β-IL-1R system is a central element of kidney inflammation and the systemic consequences. Seminal discoveries of recent years have expanded this central paradigm of inflammation. This review gives an overview of arising concepts of inflammasome and IL-1α/β regulation in renal cells and in experimental kidney disease models. There is a pipeline of compounds that can interfere with the inflammasome-IL-1α/IL-β-IL-1R system, ranging from recently described small molecule inhibitors of NLRP3, a component of the inflammasome complex, to regulatory agency-approved IL-1-neutralizing biologic drugs. Based on strong theoretic and experimental rationale, the potential therapeutic benefits of using such compounds to block the inflammasome-IL-1α/IL-β-IL-1R system in kidney disease should be further explored.
Collapse
Affiliation(s)
- Hans-Joachim Anders
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Ludwig-Maximilians Universität, Munich, Germany
| |
Collapse
|
11
|
Little MH, Kairath P. Regenerative medicine in kidney disease. Kidney Int 2016; 90:289-299. [DOI: 10.1016/j.kint.2016.03.030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 03/10/2016] [Accepted: 03/17/2016] [Indexed: 12/31/2022]
|
12
|
Purves JT, Hughes FM. Inflammasomes in the urinary tract: a disease-based review. Am J Physiol Renal Physiol 2016; 311:F653-F662. [PMID: 27170685 DOI: 10.1152/ajprenal.00607.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 05/04/2016] [Indexed: 12/28/2022] Open
Abstract
Inflammasomes are supramolecular structures that sense molecular patterns from pathogenic organisms or damaged cells and trigger an innate immune response, most commonly through production of the proinflammatory cytokines IL-1β and IL-18, but also through less understood mechanisms independent of these cytokines. Great strides have been made in understanding these structures and their dysfunction in various inflammatory diseases, lending new insights into urological and renal problems. From a clinical perspective, benign urinary pathology almost universally involves the inflammatory process, and understanding how inflammasomes translate etiological conditions (diabetes, obstruction, stones, urinary tract infections, etc.) into acute and chronic inflammatory responses is critical to understanding these diseases at a molecular level. To date, inflammasome components have been found in the bladder, prostate, and kidney and have been shown to be activated in response to several infectious and noninfectious insults. In this review, we summarize what is known regarding inflammasomes in both the upper and lower urinary tract and describe several common disease states where they potentially play critical roles.
Collapse
Affiliation(s)
- J Todd Purves
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - F Monty Hughes
- Division of Urology, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
13
|
Zhang M, Guo Y, Fu H, Hu S, Pan J, Wang Y, Cheng J, Song J, Yu Q, Zhang S, Xu JF, Pei G, Xiang X, Yang P, Wang CY. Chop deficiency prevents UUO-induced renal fibrosis by attenuating fibrotic signals originated from Hmgb1/TLR4/NFκB/IL-1β signaling. Cell Death Dis 2015; 6:e1847. [PMID: 26247732 PMCID: PMC4558499 DOI: 10.1038/cddis.2015.206] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 06/10/2015] [Accepted: 06/19/2015] [Indexed: 01/14/2023]
Abstract
Renal fibrosis, particularly tubulointerstitial fibrosis is considered to be the final manifestation of almost all chronic kidney diseases (CKDs). Herein we demonstrated evidence that CHOP-related ER stress is associated with the development of renal fibrosis in both CKD patients and unilateral ureteral obstruction (UUO)-induced animals, and specifically, mice deficient in Chop were protected from UUO-induced renal fibrosis. Mechanistic studies revealed that loss of Chop protected tubular cells from UUO-induced apoptosis and secondary necrosis along with attenuated Hmgb1 passive release and active secretion. As a result, Chop deficiency suppressed Hmgb1/TLR4/NFκB signaling, which then repressed UUO-induced IL-1β production. Consequently, the IL-1β downstream Erk1/2 activity and its related c-Jun transcriptional activity were reduced, leading to attenuated production of TGF-β1 following UUO insult. It was further noted that reduced IL-1β production also inhibited UUO-induced PI3K/AKT signaling, and both of which ultimately protected mice from UUO-induced renal fibrosis. Together, our data support that suppression of CHOP expression could be a viable therapeutic strategy to prevent renal fibrosis in patients with CKDs.
Collapse
Affiliation(s)
- M Zhang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Y Guo
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - H Fu
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - S Hu
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J Pan
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Y Wang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J Cheng
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J Song
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Q Yu
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - S Zhang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - J-F Xu
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical College, Dongguan, China
| | - G Pei
- Department of Nephrology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - X Xiang
- Department of Emergency Medicine, Institute of Emergency Medicine and Rare Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| | - P Yang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - C-Y Wang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Clinical Immunology, Institute of Laboratory Medicine, Guangdong Medical College, Dongguan, China
- Department of Emergency Medicine, Institute of Emergency Medicine and Rare Diseases, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
14
|
Thangada S, Shapiro LH, Silva C, Yamase H, Hla T, Ferrer FA. Treatment with the immunomodulator FTY720 (fingolimod) significantly reduces renal inflammation in murine unilateral ureteral obstruction. J Urol 2014; 191:1508-16. [PMID: 24679864 DOI: 10.1016/j.juro.2013.10.072] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2013] [Indexed: 02/07/2023]
Abstract
PURPOSE The S1P signaling pathway represents an important potential target for the modulation of tissue inflammation/injury. The immunomodulator FTY720, also known as fingolimod, is a potent agonist for multiple S1P receptors that was approved by the Food and Drug Administration to treat multiple sclerosis. We examined the therapeutic role of FTY720 for renal injury secondary to unilateral ureteral obstruction. MATERIALS AND METHODS CB57BL/6 mice underwent a sham procedure or unilateral ureteral obstruction and were treated with FTY720 by gavage for 1, 3 and 5 days. Control groups received vehicle. Ligated and unligated renal tissue was examined for histopathological changes, inflammatory and fibrotic markers, TGF-β1, α-SMA, and macrophage infiltration by Western blot and immunohistochemistry. Proinflammatory and profibrotic cytokines were profiled by quantitative reverse transcriptase-polymerase chain reaction. RESULTS Pathological evaluation revealed that FTY720 treatment resulted in a significant reduction in inflammatory infiltration in obstructed kidneys compared to controls. Immunohistochemical and Western blot showed that TGF-β1 and α-SMA protein levels were similarly decreased, as was macrophage infiltration into the renal interstitial space, compared to untreated mice. In agreement with these observations quantitative reverse transcriptase-polymerase chain reaction revealed that inflammatory and fibrotic cytokines (MCP-1, IL-1β, CXCL1, TNF-α and TGF-β1) were also significantly decreased in the FTY720 group. CONCLUSIONS This study suggests that in a murine ureteral obstruction model FTY720 significantly inhibited the production of inflammatory cytokines and factors regulating interstitial fibrosis and extracellular matrix accumulation. These findings were associated with decreased evidence of renal injury on pathological examination, suggesting that FTY720 or related compounds may be valuable modulators of obstruction induced renal injury.
Collapse
Affiliation(s)
- Shobha Thangada
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, Connecticut; Department of Urology, Connecticut Children's Medical Center, Hartford, Connecticut
| | - Linda H Shapiro
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, Connecticut; Department of Urology, Connecticut Children's Medical Center, Hartford, Connecticut
| | - Cynthia Silva
- Department of Nephrology, Connecticut Children's Medical Center, Hartford, Connecticut
| | - Harold Yamase
- Department of Pathology, University of Connecticut Health Center, Farmington, Connecticut
| | - Timothy Hla
- Center for Vascular Biology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
| | - Fernando A Ferrer
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, Connecticut; Department of Urology, Connecticut Children's Medical Center, Hartford, Connecticut.
| |
Collapse
|
15
|
Turner CM, Arulkumaran N, Singer M, Unwin RJ, Tam FWK. Is the inflammasome a potential therapeutic target in renal disease? BMC Nephrol 2014; 15:21. [PMID: 24450291 PMCID: PMC3918225 DOI: 10.1186/1471-2369-15-21] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 01/07/2014] [Indexed: 02/08/2023] Open
Abstract
The inflammasome is a large, multiprotein complex that drives proinflammatory cytokine production in response to infection and tissue injury. Pattern recognition receptors that are either membrane bound or cytoplasmic trigger inflammasome assembly. These receptors sense danger signals including damage-associated molecular patterns and pathogen-associated molecular patterns (DAMPS and PAMPS respectively). The best-characterized inflammasome is the NLRP3 inflammasome. On assembly of the NLRP3 inflammasome, post-translational processing and secretion of pro-inflammatory cytokines IL-1β and IL-18 occurs; in addition, cell death may be mediated via caspase-1. Intrinsic renal cells express components of the inflammasome pathway. This is most prominent in tubular epithelial cells and, to a lesser degree, in glomeruli. Several primary renal diseases and systemic diseases affecting the kidney are associated with NLRP3 inflammasome/IL-1β/IL-18 axis activation. Most of the disorders studied have been acute inflammatory diseases. The disease spectrum includes ureteric obstruction, ischaemia reperfusion injury, glomerulonephritis, sepsis, hypoxia, glycerol-induced renal failure, and crystal nephropathy. In addition to mediating renal disease, the IL-1/ IL-18 axis may also be responsible for development of CKD itself and its related complications, including vascular calcification and sepsis. Experimental models using genetic deletions and/or receptor antagonists/antiserum against the NLRP3 inflammasome pathway have shown decreased severity of disease. As such, the inflammasome is an attractive potential therapeutic target in a variety of renal diseases.
Collapse
Affiliation(s)
| | - Nishkantha Arulkumaran
- Imperial College Kidney and Transplant Institute, Hammersmith Hospital, Imperial College London, London, UK.
| | | | | | | |
Collapse
|
16
|
Carlsen I, Nilsson L, Frøkiaer J, Nørregaard R. Changes in phosphorylated heat-shock protein 27 in response to acute ureteral obstruction in rats. Acta Physiol (Oxf) 2013; 209:167-78. [PMID: 23834360 DOI: 10.1111/apha.12135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 09/18/2012] [Accepted: 06/05/2013] [Indexed: 12/19/2022]
Abstract
AIM In vivo, renal medullary interstitial cells (RMICs) and collecting duct principal cells (mpkCCD cells) are subjected to inflammatory, oxidative and mechanical stress as a result of unilateral ureteral obstruction (UUO). Because heat-shock protein (HSP) 27 and HSP70 are induced by cellular stresses and play a role in cytoprotection, we hypothesized that HSP27 and HSP70 are increased in rats subjected to acute UUO and in RMICs and mpkCCD cells exposed to inflammatory, oxidative or mechanical stress. METHODS Rats were subjected to acute UUO for 6 h and 12 h. To examine the expression of HSP27, phosphorylated HSP27 (pHSP27) and HSP70 in response to inflammatory, oxidative and mechanical stress in vitro, we exposed RMICs and mpkCCD cells to interleukin 1β (IL-1β), hydrogen peroxide (H2 O2 ), and stretch stimulation over time. RESULTS The phosphorylated form of HSP27 (pHSP27) was increased in the renal inner medulla (IM) after 6-h and 12-h UUO, while HSP27 and HSP70 were unchanged. Furthermore, after 6 h and 12 h of UUO, the expression of inflammatory (IL-1β) and oxidative [haem oxygenase 1 (HO-1)] markers was induced. Exposure to inflammatory, oxidative and mechanical stress changed HSP27 and pHSP27 expression in RMICs but not in mpkCCD cells, while HSP70 was not affected by any of the stress conditions. Exposure of RMICs to oxidative and mechanical stress induced HSP27 phosphorylation via a p38-dependent mechanism. CONCLUSION These data demonstrate that, in response to acute UUO, different forms of cellular stresses modulate HSP27 expression and phosphorylation in RMICs. This may affect the ability of renal cells to mount an effective cytoprotective response.
Collapse
Affiliation(s)
- I Carlsen
- The Water and Salt Research Center, Aarhus University, Aarhus, Denmark; Institute of Clinical Medicine, Aarhus University Hospital-Skejby, Aarhus, Denmark
| | | | | | | |
Collapse
|
17
|
Boesen EI. Chronic elevation of IL-1β induces diuresis via a cyclooxygenase 2-mediated mechanism. Am J Physiol Renal Physiol 2013; 305:F189-98. [PMID: 23657858 DOI: 10.1152/ajprenal.00075.2013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Chronic renal inflammation is an increasingly recognized phenomenon in multiple disease states, but the impact of specific cytokines on renal function is unclear. Previously, we found that 14-day interleukin-1β (IL-1β) infusion increased urine flow in mice. To determine the mechanism by which this occurs, the current study tested the possible involvement of three classical prodiuretic pathways. Chronic IL-1β infusion significantly increased urine flow (6.5 ± 1 ml/day at day 14 vs. 2.3 ± 0.3 ml/day in vehicle group; P < 0.05) and expression of cyclooxygenase (COX)-2, all three nitric oxide synthase (NOS) isoforms, and endothelin (ET)-1 in the kidney (P < 0.05 in all cases). Urinary prostaglandin E metabolite (PGEM) excretion was also significantly increased at day 14 of IL-1β infusion (1.21 ± 0.26 vs. 0.29 ± 0.06 ng/day in vehicle-infused mice; P = 0.001). The selective COX-2 inhibitor celecoxib markedly attenuated urinary PGEM excretion and abolished the diuretic response to chronic IL-1β infusion. In contrast, deletion of NOS3, or inhibition of NOS1 with L-VNIO, did not blunt the diuretic effect of IL-1β, nor did pharmacological blockade of endothelin ETA and ETB receptors with A-182086. Consistent with a primary effect on water transport, IL-1β infusion markedly reduced inner medullary aquaporin-2 expression (P < 0.05) and did not alter urinary Na⁺ or K⁺ excretion. These data indicate a critical role for COX-2 in mediating the effects of chronic IL-1β elevation on the kidney.
Collapse
Affiliation(s)
- E I Boesen
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
18
|
Cao Q, Wang Y, Harris DCH. Pathogenic and protective role of macrophages in kidney disease. Am J Physiol Renal Physiol 2013; 305:F3-11. [PMID: 23637206 DOI: 10.1152/ajprenal.00122.2013] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Macrophages (MΦ) are located throughout kidney tissue, where they play important roles in homeostasis, surveillance, tolerance, and cytoprotection. MΦ are highly heterogeneous cells and exhibit distinct phenotypic and functional characteristics depending on their microenvironment and the disease type and stage. Recent studies have identified a dual role for MΦ in several murine models of kidney disease. In this review, we discuss the pathogenic and protective roles of the various MΦ subsets in experimental and human kidney diseases and summarize current progress toward the therapeutic use of MΦ in kidney diseases.
Collapse
Affiliation(s)
- Qi Cao
- Centre for Transplantation and Renal Research, Westmead Millennium Institute, University of Sydney, Darcy Rd., Westmead, Sydney, NSW, Australia.
| | | | | |
Collapse
|
19
|
Abstract
Renal inflammation is a universal response to infectious and noninfectious triggers. Sensors of the innate immune system, such as Toll-like receptors or RIG-like receptors, provide danger recognition platforms on renal cells that integrate and translate the diverse triggers of renal inflammation by inducing cell activation and the secretion of proinflammatory cytokines and chemokines. As a new entry, the inflammasome-forming NLR genes integrate various danger signals into caspase-1-activating platforms that regulate the processing and secretion of pro-IL-1β and pro-IL-18 into the mature and active cytokines. Accumulating data now document a role for the NLRP3 inflammasome and IL-1β/IL-18 in many diseases, including atherosclerosis, diabetes, amyloidosis, malaria, crystal-related diseases, and other autoinflammatory disorders, identifying this innate immune pathway as an attractive therapeutic target. Here we review the current knowledge regarding inflammasome signaling and outline existing evidence on the expression and functional role of the inflammasome-caspase-1-IL-1β/IL-18 axis in kidney disease. We further provide a perspective on the potential roles of the inflammasomes in the pathogenesis of acute and chronic kidney diseases.
Collapse
|
20
|
|
21
|
Macrophages expressing heme oxygenase-1 improve renal function in ischemia/reperfusion injury. Mol Ther 2010; 18:1706-13. [PMID: 20551909 DOI: 10.1038/mt.2010.100] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Acute kidney injury has a high mortality and lacks specific therapies, with ischemia/reperfusion injury (IRI) being the predominant cause. Macrophages (M phi) have been used successfully in cell therapy to deliver targeted therapeutic genes in models of inflammatory kidney disease. Heme oxygenase-1 (HO-1) catalyzes heme breakdown and has important cytoprotective functions. We hypothesized that administration of M phi modified to overexpress HO-1 would protect from renal IRI. Using an adenoviral construct (Ad-HO-1), HO-1 was overexpressed in primary bone marrow-derived M phi (BMDM). In vitro Ad-HO-1 M phi showed an anti-inflammatory phenotype with increased phagocytosis of apoptotic cells (ACs) and increased interleukin (IL)-10 but reduced TNF-alpha and nitric oxide (NO) following lipopolysaccharide/interferon-gamma (IFN gamma) stimulation compared to control transduced or unmodified M phi. In vivo, intravenously (IV) injected M phi homed preferentially to the post-IRI kidney compared to uninjured control following experimental IRI. At 24 hours postinjury, despite equivalent levels of tubular necrosis, apoptosis, and capillary density between groups, the injection of Ad-HO-1 M phi resulted in preserved renal function (serum creatinine reduced by 46%), and reduced microvascular platelet deposition. These data demonstrate that genetically modified M phi improve the outcomes in IRI when administered after the establishment of structural injury, raising the prospect of targeted cell therapy to support the function of the acutely injured kidney.
Collapse
|
22
|
|
23
|
Grande MT, Pérez-Barriocanal F, López-Novoa JM. Role of inflammation in túbulo-interstitial damage associated to obstructive nephropathy. JOURNAL OF INFLAMMATION-LONDON 2010; 7:19. [PMID: 20412564 PMCID: PMC2873503 DOI: 10.1186/1476-9255-7-19] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 04/22/2010] [Indexed: 02/07/2023]
Abstract
Obstructive nephropathy is characterized by an inflammatory state in the kidney, that is promoted by cytokines and growth factors produced by damaged tubular cells, infiltrated macrophages and accumulated myofibroblasts. This inflammatory state contributes to tubular atrophy and interstitial fibrosis characteristic of obstructive nephropathy. Accumulation of leukocytes, especially macrophages and T lymphocytes, in the renal interstitium is strongly associated to the progression of renal injury. Proinflammatory cytokines, NF-κB activation, adhesion molecules, chemokines, growth factors, NO and oxidative stress contribute in different ways to progressive renal damage induced by obstructive nephropathy, as they induce leukocytes recruitment, tubular cell apoptosis and interstitial fibrosis. Increased angiotensin II production, increased oxidative stress and high levels of proinflammatory cytokines contribute to NF-κB activation which in turn induce the expression of adhesion molecules and chemokines responsible for leukocyte recruitment and iNOS and cytokines overexpression, which aggravates the inflammatory response in the damaged kidney. In this manuscript we revise the different events and regulatory mechanisms involved in inflammation associated to obstructive nephropathy.
Collapse
Affiliation(s)
- María T Grande
- Instituto "Reina Sofía" de Investigación Nefrológica, Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca, Spain.
| | | | | |
Collapse
|
24
|
López-Novoa JM, Nieto MA. Inflammation and EMT: an alliance towards organ fibrosis and cancer progression. EMBO Mol Med 2009; 1:303-14. [PMID: 20049734 PMCID: PMC3378143 DOI: 10.1002/emmm.200900043] [Citation(s) in RCA: 531] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Revised: 08/17/2009] [Accepted: 08/26/2009] [Indexed: 12/13/2022] Open
Abstract
Recent advances in our understanding of the molecular pathways that govern the association of inflammation with organ fibrosis and cancer point to the epithelial to mesenchymal transition (EMT) as the common link in the progression of these devastating diseases. The EMT is a crucial process in the development of different tissues in the embryo and its reactivation in the adult may be regarded as a physiological attempt to control inflammatory responses and to 'heal' damaged tissue. However, in pathological contexts such as in tumours or during the development of organ fibrosis, this healing response adopts a sinister nature, steering these diseases towards metastasis and organ failure. Importantly, the chronic inflammatory microenvironment common to fibrotic and cancer cells emerges as a decisive factor in the induction of the pathological EMT.
Collapse
Affiliation(s)
- Jose Miguel López-Novoa
- Departamento de Fisiología y Farmacología, Universidad de Salamanca, Edificio Departamental, Campus Miguel de UnamunoSalamanca, Spain
| | - M Angela Nieto
- Instituto de Neurociencias CSIC-UMHSan Juan de Alicante, Spain
| |
Collapse
|
25
|
Matsumoto K, Fukuda N, Abe M, Fujita T. Dendritic cells and macrophages in kidney disease. Clin Exp Nephrol 2009; 14:1-11. [PMID: 19688180 DOI: 10.1007/s10157-009-0218-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2008] [Accepted: 07/09/2009] [Indexed: 01/25/2023]
Affiliation(s)
- Koichi Matsumoto
- Division of Nephrology, Hypertension and Endocrinology, Department of Internal Medicine, Nihon University School of Medicine, 30-1 Oyaguchi-Kami-Machi, Itabashi-ku, Tokyo, 173-8610, Japan.
| | | | | | | |
Collapse
|
26
|
Johansson ÅC, Nandakumar KS, Persson AM, Olsson I, Hansson M. Secretory lysosome targeting and induced secretion of human soluble TNF-α receptor in murine hematopoietic cells in vivo as a principle for immunoregulation in inflammation and malignancy. Exp Hematol 2009; 37:969-78. [DOI: 10.1016/j.exphem.2009.05.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2009] [Revised: 05/11/2009] [Accepted: 05/22/2009] [Indexed: 11/30/2022]
|
27
|
Jones LK, O’Sullivan KM, Semple T, Kuligowski MP, Fukami K, Ma FY, Nikolic-Paterson DJ, Holdsworth SR, Kitching AR. IL-1RI deficiency ameliorates early experimental renal interstitial fibrosis. Nephrol Dial Transplant 2009; 24:3024-32. [DOI: 10.1093/ndt/gfp214] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
28
|
Dong X, Bachman LA, Miller MN, Nath KA, Griffin MD. Dendritic cells facilitate accumulation of IL-17 T cells in the kidney following acute renal obstruction. Kidney Int 2008; 74:1294-309. [PMID: 18974760 DOI: 10.1038/ki.2008.394] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Acute urinary obstruction causes interstitial inflammation with leukocyte accumulation and the secretion of soluble mediators. Here we show that unilateral ureteral ligation caused a progressive increase in renal F4/80(+) and F4/80(-) dendritic cells, monocytes, neutrophils and T-cells 24-72 h following obstruction. Depletion of dendritic cells by clodronate pretreatment showed these cells to be the most potent source of tumor necrosis factor and other pro-inflammatory mediators in the obstructed kidney. F4/80(+) dendritic cells and T-cells co-localized in the cortico-medullary junction and cortex of the obstructed kidney. Cytokine secretion patterns and surface phenotypes of T-cells from obstructed kidneys were found to include interferon-gamma-secreting CD4(+) and CD8(+) memory T-cells as well as interleukin 17 (IL-17)-secreting CD4(+) memory T-cells. Depletion of the intra-renal dendritic cells prior to ligation did not numerically reduce T-cells in obstructed kidneys but attenuated interferon-gamma and IL-17-competent T-cells. Our study shows that intra-renal dendritic cells are a previously unidentified early source of proinflammatory mediators after acute urinary obstruction and play a specific role in recruitment and activation of effector-memory T-cells including IL-17-secreting CD4(+) T-cells.
Collapse
Affiliation(s)
- Xiangyang Dong
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | | | | | | | | |
Collapse
|
29
|
YAMAGISHI H, UTSUNOMIYA Y, YOKOO T, KAWAMURA T, HOSOYA T. Use of genetically modified bone marrow-derived vehicle cells to deliver anti-inflmamatory cytokines to inflamed interstitium. Nephrology (Carlton) 2008. [DOI: 10.1111/j.1440-1797.2002.tb00550.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
30
|
Abstract
Renal inflammation may result from a myriad of insults and often is characterized by the presence of infiltrating inflammatory leukocytes within the glomerulus or tubulointerstitium. Accumulating evidence indicates that infiltrating leukocytes are key players in the induction of renal injury. Although renal inflammation often is followed by the development of fibrosis with loss of renal function, it can resolve. Resolution may be spontaneous as in poststreptococcal glomerulonephritis or after the administration of effective treatment such as immunosuppressive agents. The mechanisms and cells underlying the resolution process and the exact temporal sequence remains uncertain at present but likely involves the removal of injurious leukocytes, the down-regulation of immune responses, and the alteration of the phenotype of infiltrating macrophages from proinflammatory to prorepair. In this review we examine the role of leukocytes in both renal inflammation and repair.
Collapse
Affiliation(s)
- David Ferenbach
- MRC Centre for Inflammation Research, University of Edinburgh, Queens Medical Research Centre, Edinburgh, United Kingdom
| | | | | |
Collapse
|
31
|
Abstract
Macrophages are inflammatory cells with important roles in the propagation of renal injury. More recently they have also been shown to be important in the resolution of inflammation. Wang et al. show that macrophages can be modulated ex vivo by cytokine stimulation, localize to renal tissue, and slow progression of experimental glomerular inflammation. Thus strategies targeting macrophage function have considerable therapeutic potential.
Collapse
Affiliation(s)
- D C Kluth
- Medical Research Council Centre for Inflammation Research, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, United Kingdom.
| |
Collapse
|
32
|
Wang Y, Wang YP, Zheng G, Lee VWS, Ouyang L, Chang DHH, Mahajan D, Coombs J, Wang YM, Alexander SI, Harris DCH. Ex vivo programmed macrophages ameliorate experimental chronic inflammatory renal disease. Kidney Int 2007; 72:290-299. [PMID: 17440493 DOI: 10.1038/sj.ki.5002275] [Citation(s) in RCA: 286] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Macrophage infiltration of the kidney is a prominent feature associated with the severity of renal injury and progressive renal failure. To determine the influence of macrophages in renal disease models in the absence of endogenous T and B cells, we performed adoptive transfer of macrophages into severe combined immunodeficient (SCID) mice. In this study, macrophages were isolated from the spleens of BALB/c mice and stimulated with lipopolysaccharide to induce classically activated M1 macrophages or with interleukin-4 (IL-4) and IL-13 to induce alternatively activated M2 macrophages. These macrophages were then infused into SCID mice with adriamycin nephropathy; an in vivo model of chronic inflammatory renal disease analogous to human focal segmental glomerulosclerosis. Mice infused with M1 macrophages had a more severe histological and functional injury, whereas M2 macrophage-induced transfused mice had reduced histological and functional injury. Both M1 and M2 macrophages localized preferentially to the area of injury and maintained their phenotypes even after 4 weeks. The protective effect of M2 macrophages was associated with reduced accumulation and possibly downregulated chemokine and inflammatory cytokine expression of the host infiltrating macrophages. Our findings demonstrate that macrophages not only act as effectors of immune injury but can be induced to provide protection against immune injury.
Collapse
Affiliation(s)
- Y Wang
- Centre for Transplantation and Renal Research, The University of Sydney at Westmead Millennium Institute, Westmead, New South Wales, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Isaka Y. Gene therapy targeting kidney diseases: routes and vehicles. Clin Exp Nephrol 2006; 10:229-35. [PMID: 17186326 DOI: 10.1007/s10157-006-0442-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Accepted: 10/02/2006] [Indexed: 11/24/2022]
Abstract
Renal gene therapy may offer new strategies to treat diseases of native and transplanted kidneys. Several experimental techniques have been developed and employed using nonviral, viral, and cellular vectors. The most efficient viral vector for in vivo transfection appears to be adenovirus. In addition, enhanced naked plasmid techniques, such as the hemagglutinating virus of Japan (HVJ)-liposome method, electroporation, the hydrodynamic method, and ultrasound with microbubbles, are promising. Trapping genetically modified macrophages in the inflamed kidneys is an elegant method for site-specific gene delivery. The choice of delivery vehicle as well as the administration route determines the site of transduction. In conclusion, for both in vivo and ex vivo renal transfection, enhanced naked plasmids, adenoviruses, and modified cell vectors offer the best prospects for effective clinical application. Moreover, the development of safer and nonimmunogenic vectors may realize clinical renal gene therapy in the near future.
Collapse
Affiliation(s)
- Yoshitaka Isaka
- Department of Advanced Technology for Transplantation, Osaka University Graduate School of Medicine, Suita 565-0871, Japan.
| |
Collapse
|
34
|
Chevalier RL. Obstructive nephropathy: towards biomarker discovery and gene therapy. ACTA ACUST UNITED AC 2006; 2:157-68. [PMID: 16932414 DOI: 10.1038/ncpneph0098] [Citation(s) in RCA: 143] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2005] [Accepted: 12/05/2005] [Indexed: 12/16/2022]
Abstract
Obstructive nephropathy is a major cause of renal failure, particularly in infants and children. Cellular and molecular mechanisms responsible for the progression of the tubular atrophy and interstitial fibrosis-processes that lead to nephron loss-have been elucidated in the past 5 years. Following urinary tract obstruction and tubular dilatation, a cascade of events results in upregulation of the intrarenal renin-angiotensin system, tubular apoptosis and macrophage infiltration of the interstitium. This is followed by accumulation of interstitial fibroblasts through proliferation of resident fibroblasts and epithelial-mesenchymal transformation of renal tubular cells. Under the influence of cytokines, chemokines and other signaling molecules produced by tubular and interstitial cells, fibroblasts undergo transformation to myofibroblasts that induce expansion of the extracellular matrix. The cellular interactions that regulate development of interstitial inflammation, tubular apoptosis and interstitial fibrosis are complex. Changes in renal gene expression and protein production afford many potential biomarkers of disease progression and targets for therapeutic manipulation. These include signaling molecules and receptors involved in macrophage recruitment and proliferation, tubular death signals and survival factors, and modulators of epithelial-mesenchymal transformation. Targeted gene deletion and various forms of gene therapy have been used in experimental obstructive nephropathy, mostly rodent models of unilateral ureteral obstruction or cell culture techniques. Further refinement of these models is needed to develop a matrix of biomarkers with clinical predictive value, as well as molecular therapies that will prevent or reverse the renal structural and functional consequences of obstructive nephropathy.
Collapse
Affiliation(s)
- Robert L Chevalier
- Department of Pediatrics at the University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
35
|
Hughes J. Leukocytes in tubulointerstitial inflammation. Kidney Int 2006; 69:8-10. [PMID: 16374414 DOI: 10.1038/sj.ki.5000077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The work of Lange-Sperandio et al in this issue explores the differential role of beta2 integrins in promoting the macrophage infiltration characteristic of the obstructed kidneys of neonatal mice. Future work is needed to define factors that regulate macrophage death within or emigration from the kidney as well as to explore strategies to modulate macrophage phenotype. This knowledge will assist the development of novel therapeutic agents to limit injury and promote tissue repair.
Collapse
Affiliation(s)
- J Hughes
- Medical Research Council Centre for Inflammation Research, The Queen's Medical Research Institute, Edinburgh, UK.
| |
Collapse
|
36
|
Lange-Sperandio B, Schimpgen K, Rodenbeck B, Chavakis T, Bierhaus A, Nawroth P, Thornhill B, Schaefer F, Chevalier RL. Distinct roles of Mac-1 and its counter-receptors in neonatal obstructive nephropathy. Kidney Int 2006; 69:81-8. [PMID: 16374427 DOI: 10.1038/sj.ki.5000017] [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: 02/07/2023]
Abstract
Urinary tract obstruction during renal development leads to tubular atrophy and interstitial fibrosis. Inflammatory macrophages are crucial in this process, and beta2-integrins play a major role in leukocyte recruitment. We investigated the role of beta2-integrins and their major counter-receptors (intercellular adhesion molecule-1 (ICAM-1), receptor for advanced glycation endproducts (RAGE), junctional adhesion molecule (JAM)-C) in obstructive nephropathy in neonatal mice. Two-day-old beta2-integrin-deficient mice (Mac-1-/- and LFA-1-/-(deficient for leukocyte function-associated antigen-1)) and wild-type mice (C57BL/6) underwent unilateral ureteral obstruction (UUO) or sham operation. After 1, 5 or 12 days of obstruction, renal macrophage infiltration and tubulointerstitial damage were quantitated. Tissue abundance of Mac-1 and its ligands ICAM-1, RAGE and JAM-C was examined by Western blot and immunoprecipitation. Deficiency of either integrin was associated with reduced early macrophage invasion into the obstructed kidney. After 12 days of UUO, macrophage infiltration and tubulointerstitial injury were reduced only in Mac-1-/- but not in LFA-1-/- mice. Besides ICAM-1, an upregulation of two novel Mac-1 ligands, RAGE and JAM-C were observed, however, with distinct time courses. We conclude that beta2-integrins mediate macrophage infiltration in UUO. Mac-1 is the predominant leukocyte integrin involved in leukocyte recruitment after obstruction. ICAM-1 and its new ligands RAGE and JAM-C are sequentially activated in UUO. Blocking of Mac-1 and its ligands may confer synergistic renoprotective effects in neonatal obstructive nephropathy.
Collapse
Affiliation(s)
- B Lange-Sperandio
- Department of Pediatrics, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Docherty NG, O'Sullivan OE, Healy DA, Fitzpatrick JM, Watson RWG. Evidence that inhibition of tubular cell apoptosis protects against renal damage and development of fibrosis following ureteric obstruction. Am J Physiol Renal Physiol 2006; 290:F4-13. [PMID: 16339963 DOI: 10.1152/ajprenal.00045.2005] [Citation(s) in RCA: 137] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Ureteric obstruction is frequently encountered in primary care urology and can lead to damage to the ipsilateral kidney. Relief of all types of obstruction generally leads to the normalization of any deterioration in renal function noted at diagnosis. However, some evidence from animal models suggests that obstruction can cause progressive deleterious effects on renal function and blood pressure control, especially in the presence of preexisting pathologies such as essential hypertension. The last 10 years have seen a proliferation of studies in rodents wherein complete unilateral ureteric obstruction has been used as a model of renal fibrosis. However, the relevance of the findings to human obstructive uropathy has, in many cases, not been the primary aim. In this review, we outline the major events linking damage to the renal parenchyma and cell death to the evolution of fibrosis following obstruction. Special focus is given to the role of apoptosis as a major cause of cell death during and post-complete ureteric obstruction. Several interventions that reduce tubular apoptosis are discussed in terms of their ability to prevent subsequent progression to end-organ damage and fibrosis.
Collapse
Affiliation(s)
- Neil G Docherty
- Department of Surgery, Conway Institute of Biomolecular and Biomedical Sciences, Univ. College Dublin, Belfield, Dublin 4, Republic of Ireland
| | | | | | | | | |
Collapse
|
38
|
Ren L, Blanchette JB, White LR, Clark SA, Heffner DJ, Tibbles LA, Muruve DA. Soluble fibronectin induces chemokine gene expression in renal tubular epithelial cells. Kidney Int 2005; 68:2111-20. [PMID: 16221210 DOI: 10.1111/j.1523-1755.2005.00667.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Increasing proteinuria in kidney disease is associated with an increased risk of renal failure. Urinary proteins such as albumin induce inflammatory signaling and gene expression in tubular epithelial cells (TECs). Fibronectin is an extracellular matrix protein that can exist in soluble form and is excreted in the urine of patients with glomerular disease. METHODS To explore the impact of soluble fibronectin on tubular epithelium, murine TECs were stimulated with soluble fibronectin and chemokine mRNA was determined by RNase protection assay. RESULTS Fibronectin induced the expression of inflammatory chemokine genes, including monocyte chemoattractant protein-1 (MCP-1) (CCL2) and macrophage inflammatory protein-2 (MIP-2) within 2 hours in a dose-dependent manner. Phosphorylation of Src family tyrosine kinases was also increased in TECs following exposure to fibronectin. Src tyrosine kinases were involved in the fibronectin activation of MCP-1 since the Src inhibitors SU6656 and PP2 effectively reduced the induction of this chemokine. Fibronectin also induced the phosphorylation of extracellular signal-regulated protein kinase (ERK1/2) within minutes in TECs. The ERK kinase (MEK1/2) inhibitor U0126 inhibited the fibronectin induction of MCP-1 mRNA suggesting that ERK1/2 was also involved in this inflammatory pathway. Furthermore, fibronectin also induced phosphorylation of IkappaBalpha within 20 minutes in TECs. The nuclear factor-kappaB (NF-kappaB) inhibitors N-acetyl-L-cysteine (NAC) and pyrrolidinecarbodithioic acid (PDTC) effectively blocked fibronectin induction of MCP-1 mRNA. CONCLUSION Soluble fibronectin activates MCP-1 gene expression in TECs via Src tyrosine kinases, ERK1/2 and NF-kappaB. These data provide further support to the concept that proteinuria per se contributes to the tubulointerstitial injury observed in glomerular disease.
Collapse
Affiliation(s)
- Li Ren
- Division of Nephrology, Department of Medicine, University of Calgary, Alberta, Canada
| | | | | | | | | | | | | |
Collapse
|
39
|
Abstract
Macrophages and progressive tubulointerstitial disease. In chronic renal disease, tubulointerstitial inflammation and injury is associated with infiltrating macrophages. As a consequence of primary injury, proteinuria, chronic hypoxia, and glomerular-derived cytokines may all differentially modulate the expression of factors that promote macrophage recruitment. In addition to adhesion molecules and chemokines, products of complement system and renin-angiotensin system activation may direct this process. Once present at interstitial sites, macrophages interact with resident cells and extracellular matrix to generate a proinflammatory microenvironment that amplifies tissues injury and promotes scarring. There is now increasing evidence for the efficacy of interventions directed against factors that recruit, activate, or are produced by macrophages. A detailed understanding of the biology of this area may lead to the further development of therapies that will improve the outcome of renal disease.
Collapse
Affiliation(s)
- Kevin Sean Eardley
- Department of Nephrology, University Hospital Birmingham NHS Trust, Queen Elizabeth Hospital, Birmingham, United Kingdom
| | | |
Collapse
|
40
|
Abstract
There has been considerable focus on the ability of bone marrow-derived cells to differentiate into non-haematopoietic cells of various tissue lineages, including cells of the kidney. This growing evidence has led to a reconsideration of the source of cells contributing to renal repair following injury. The kidney has an inherent ability for recovery and regeneration following acute damage. It is thought that dedifferentiation of glomerular and tubular cells to a more embryonic/mesenchymal phenotype represent key processes for recovery in response to damage. However, there has been much contention as to the source of regenerating renal cells. The present review focuses on new aspects of the plasticity of intrinsic renal cells and their role in renal remodelling and scarring. Growing support also suggests that bone marrow-derived cells have the ability to contribute to structural and functional repair following acute renal failure. Evidence for bone marrow cell engraftment in the repairing kidney leading to incorporation into a variety of tissue types is discussed. Because cell death and fibrosis is a common end-point in a variety of acute and chronic renal nephropathies, the paradigm of stem cell plasticity may have important implications in the cellular and pathological mechanisms of renal injury and repair. A better understanding of the processes controlling extra-renal cell engraftment and intrinsic renal cell differentiation may provide important clues for the development of new cell-based therapies in the field of renal reparative medicine.
Collapse
Affiliation(s)
- Sharon D Ricardo
- Monash Immunology and Stem Cell Laboratories (MISCL) and Department of Anatomy and Cell Biology, Monash University, Melbourne, Victoria, Australia.
| | | |
Collapse
|
41
|
Alderuccio F, Toh BH. Induction of tolerance to self-antigens using genetically modified bone marrow cells. Expert Opin Biol Ther 2005; 4:1007-14. [PMID: 15268669 DOI: 10.1517/14712598.4.7.1007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The challenge of finding a lasting cure for autoimmune disease(s) has not been met. Although the use of systemic anti-inflammatory agents still dominates the treatment of these diseases, there is a push towards developing novel and more specific strategies. In addressing autoimmunity, there is the intrinsic need to understand the mechanisms that lead to the development and maintenance of immunological tolerance to self-antigens. Experimental evidence has shown that directed antigen expression in the thymus can induce immunological tolerance to that antigen. This forms the cornerstone of one strategy directed towards the cure of autoimmunity. In this strategy, individuals with autoimmune disease are transplanted with bone marrow stem cells that have been genetically modified and in this way allow expression of the self-antigen in the thymus.
Collapse
Affiliation(s)
- Frank Alderuccio
- Department of Immunology, Central and Eastern Clinical School, Monash University, Commercial Road, Prahran, Melbourne, Australia, 3181.
| | | |
Collapse
|
42
|
Worgall S. A realistic chance for gene therapy in the near future. Pediatr Nephrol 2005; 20:118-24. [PMID: 15549408 DOI: 10.1007/s00467-004-1680-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2004] [Revised: 08/18/2004] [Accepted: 09/08/2004] [Indexed: 10/26/2022]
Abstract
The expanding knowledge of the genetic and cellular mechanisms of human diseases in the post-genomic era coupled with the development of different vector systems to efficiently transfer genes to a variety of cell types and organs in vivo gave rise to the concept of gene therapy as a promising therapeutic option for genetic and acquired diseases. Gene therapy has been the focus of both enthusiasm and critique in the past years. Major progress has been achieved in evaluating gene therapy in clinical trials. However, a number of hurdles must still be overcome to make gene therapy safe and applicable for human diseases. Increased knowledge of the interaction of the gene therapy vehicles with the host has resulted in modifications of existing and the development of new vector systems, as well as adjustments of future clinical applications. Adeno-associated virus vectors, retrovirus- and lentivirus-based vectors show great promise for the correction of monogenic diseases. Correction of the genetic defect can be attempted by either in vivo administration to directly target a diseased organ or by administration of ex vivo genetically modified cells, e.g., bone marrow stem cells. The lack of persistent expression and the immune responses of the host have limited the use of adenovirus vectors for the permanent correction of monogenic diseases. However, the ease of production and the number of cell types and organs that can be efficiently infected make adenovirus-based vectors a promising tool for applications where permanent gene expression is not the therapeutic goal or where the induction of immune responses is the desired response, as for genetic vaccines. Overall, gene therapy remains promising for the correction of genetic as well as acquired disorders, where permanent or transient expression of a gene product will be therapeutic.
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW Macrophage infiltration is a hallmark of all forms of inflammatory and non-inflammatory renal injury. However, the classical view of macrophages as cells that cause injury has been superseded with evidence of their heterogeneous role, i.e. with involvement in all stages of the inflammatory process including tissue repair and healing. This review summarizes the major advances in macrophage biology achieved in the last year, highlighting the different activation states, how these are regulated, and their relevance in renal disease. RECENT FINDINGS New concepts have emerged concerning the factors controlling monocyte recruitment into inflamed tissue and their subsequent differentiation into activated macrophages. There is now compelling evidence for the heterogeneity of macrophages in clinical disease, i.e. they appear to be able to both promote and downregulate inflammation. An increased understanding of the factors regulating the expression of pro-inflammatory or reparative characteristics by macrophages is establishing how their function can be manipulated to attenuate renal inflammation in experimental models. SUMMARY An understanding of the role of macrophages at different time-points in renal inflammation, and the development of techniques for modulating macrophage activation in vivo, will provide a powerful method for exploiting the reparative attributes of these cells in clinical settings, restoring regulation to the inflammatory process and promoting healing.
Collapse
Affiliation(s)
- Heather M Wilson
- Department of Medicine and Therapeutics, University of Aberdeen, Institute of Medical Sciences, Foresterhill, Aberdeen AB25 2ZD, UK.
| | | | | |
Collapse
|
44
|
Abstract
Macrophage infiltration is a common feature of renal disease and their presence has been synonymous with tissue damage and progressive renal failure. More recently work has focused on the heterogeneity of macrophage activation and in particular their ability to curtail inflammation and restore normal function. This has led to the view that it is macrophage function rather than their number that is important in determining the outcome of inflammatory disease. This review will focus on the pathways that regulate macrophage infiltration and activation and how these could be manipulated to control renal inflammatory disease. In particular, the ability of specific cell surface receptors and intracellular signaling pathways to control macrophage activation and how macrophages can be genetically manipulated to develop properties that favor resolution over ongoing injury.
Collapse
Affiliation(s)
- David C Kluth
- MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, Scotland, United Kingdom.
| | | | | |
Collapse
|
45
|
van der Wouden EA, Sandovici M, Henning RH, de Zeeuw D, Deelman LE. Approaches and methods in gene therapy for kidney disease. J Pharmacol Toxicol Methods 2004; 50:13-24. [PMID: 15233963 DOI: 10.1016/j.vascn.2004.03.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2003] [Accepted: 03/08/2004] [Indexed: 11/22/2022]
Abstract
Renal gene therapy may offer new strategies to treat diseases of native and transplanted kidneys. Several experimental techniques have been developed and employed using nonviral, viral, and cellular vectors. The most efficient vector for in vivo transfection appears to be adenovirus. Glomeruli, blood vessels, interstitial cells, and pyelum can be transfected with high efficiency. In addition, electroporation and microbubbles with ultrasound, both being enhanced naked plasmid techniques, offer good opportunities. Trapping of mesangial cells into the glomeruli as well as natural targeting of monocytes or macrophages to inflamed kidneys are elegant methods for site-specific delivery of genes. For gene therapy in kidney transplantation, hemagglutinating virus of Japan liposomes are efficient vectors for tubular transfection, whereas enhanced naked plasmid techniques are suitable for glomerular transfection. However, adenovirus offers the best opportunities in a renal transplantation setup because varying parameters of graft perfusion allows targeting of different cell types. In renal grafts, lymphocytes can be used for selective targeting to sites of inflammation. In conclusion, for both in vivo and ex vivo renal transfection, enhanced naked plasmids and adenovirus offer the best perspectives for effective clinical application. Moreover, the development of safer, nonimmunogenic vectors and the large-scale production could make clinical renal gene therapy a realistic possibility for the near future.
Collapse
Affiliation(s)
- Els A van der Wouden
- Department of Clinical Pharmacology, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | | | | | | | | |
Collapse
|
46
|
Chatziantoniou C, Boffa JJ, Tharaux PL, Flamant M, Ronco P, Dussaule JC. Progression and regression in renal vascular and glomerular fibrosis. Int J Exp Pathol 2004; 85:1-11. [PMID: 15113389 PMCID: PMC2517455 DOI: 10.1111/j.0959-9673.2004.00376.x] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2004] [Accepted: 02/19/2004] [Indexed: 01/13/2023] Open
Abstract
End-stage renal disease (ESRD) is characterized by the development of fibrotic lesions in the glomerular, interstitial and vascular compartments. Renal fibrogenesis, a common complication of diabetes and hypertension, is a complex dynamic process involving several players such as inflammatory agents, cytokines, vasoactive agents and enzymes participating in extracellular matrix assembly, anchoring or degradation. The only available treatment today against chronic renal failure is dialysis or kidney transplantation, making thus ESRD one of the most expensive diseases to treat on a per-patient basis. An emerging challenge for clinicians, maybe the nephrologist's Holy Grail in the 21st century, is to stop definitively the decline of renal function and, if possible, to achieve regression of renal fibrosis and restoration of renal structure. Over the last 5 years, different approaches have been tested in experimental models of nephropathy with variable degree of success. In this review, we will focus on the mechanisms of the hypertension-associated fibrosis and the few recent studies that gave promising results for a therapeutic intervention.
Collapse
|
47
|
|
48
|
Silverstein DM, Travis BR, Thornhill BA, Schurr JS, Kolls JK, Leung JC, Chevalier RL. Altered expression of immune modulator and structural genes in neonatal unilateral ureteral obstruction. Kidney Int 2003; 64:25-35. [PMID: 12787392 DOI: 10.1046/j.1523-1755.2003.00067.x] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Congenital obstructive nephropathy is a condition characterized by hydronephrosis, tubular dilatation, apoptosis, and atrophy, as well as interstitial cellular infiltration and progressive interstitial fibrosis. The renal consequences of chronic unilateral ureteral obstruction (UUO) in the neonatal rat are similar to those of clinical congenital obstructive nephropathy. METHODS To define alterations in renal gene expression induced by chronic neonatal UUO, Sprague-Dawley rats were subjected to UUO or sham operation within the first 2 days of life, and kidneys were harvested after 12 days. RESULTS Microarray analysis revealed that the mRNA expression of multiple immune modulators, including krox24, interferon-gamma regulating factor-1 (IRF-1), monocyte chemoattractant protein-1 (MCP-1), interleukin-1beta (IL-1beta), CCAAT/enhancer binding protein (C/EBP), p21, c-fos, c-jun, and pJunB, was significantly increased in obstructed compared to sham-operated kidneys (all P < 0.05). Western blot analysis revealed significant changes in immune modulator protein abundance in the obstructed versus sham-operated kidney for krox24 (P = 0.0004), IRF-1 (P = 0.005), MCP-1 (P = 0.01), and JunD (P = 0.0008). Alternatively, the abundance of all of the immune modulator proteins was similar in sham-operated and obstructed kidneys in rats subjected to acute (4 days) neonatal UUO. Microarray analysis studies also reveal that structural genes that comprise the cytoskeleton and cell matrix are significantly up-regulated by chronic neonatal UUO, including calponin, desmin, dynamin, and lumican (all P < 0.05). CONCLUSION Multiple genes are aberrantly expressed in the kidney of rats subjected to chronic neonatal UUO. Elucidation of these genes involved in neonatal UUO may lead to new insight about congenital obstructive nephropathy.
Collapse
Affiliation(s)
- Douglas M Silverstein
- Division of Nephrology, Department of Pediatrics, Gene Therapy Program, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70124, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Wilson HM, Stewart KN, Brown PAJ, Anegon I, Chettibi S, Rees AJ, Kluth DC. Bone-marrow-derived macrophages genetically modified to produce IL-10 reduce injury in experimental glomerulonephritis. Mol Ther 2002; 6:710-7. [PMID: 12498767 DOI: 10.1006/mthe.2002.0802] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Macrophages are intimately involved in the development of immune-mediated inflammation, including glomerulonephritis. We have transduced primary cultures of macrophages to express IL-10 and tested the ability of these cells to control rat nephrotoxic nephritis (NTN), a model of human glomerulonephritis. Ad-IL-10-transduced bone-marrow-derived macrophages (BMDM) produced large amounts of IL-10 in culture, and their TNF-alpha production was decreased in response to interferon-gamma and LPS. Transduced macrophages were injected into the renal artery of rats, 6 h after the induction of NTN, where they localized efficiently to inflamed rat glomeruli. Delivery of IL-10-expressing macrophages to nephritic rats produced a marked reduction in albuminuria compared with unmodified NTN or injection of Ad-null-transduced BMDM. IL-10 treatment decreased the number of glomerular ED1- and ED3-positive cells, MHC class II expression, and the number of fibrinoid lesions. Interestingly, anti-inflammatory changes in the Ad-IL-10-injected kidney were mirrored by changes in the contralateral kidney. These results highlight that Ad-IL-10-transduced macrophages infiltrate inflamed glomeruli and reduce the severity of glomerular inflammation, emphasizing the value of local delivery of genetically modified macrophages in the manipulation of inflammatory disease.
Collapse
Affiliation(s)
- Heather M Wilson
- Department of Medicine and Therapeutics, Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Interstitial fibrosis has a major role in the progression of renal diseases. Several animal models are available for the study of renal fibrosis. The models of aminonucleoside-induced nephrotic syndrome, cyclosporin nephrotoxicity, and passive Heyman nephritis are characterized by molecular and cellular events similar to those that occur in obstructive nephropathy. Additionally, inhibition of angiotensin-converting enzyme exerts salutary effects on the progression of renal fibrosis in obstructive nephropathy. Unilateral ureteral obstruction (UUO) has emerged as an important model for the study of the mechanisms of renal fibrosis and also for the evaluation of the impact of potential therapeutic approaches to ameliorate renal disease. Many quantifiable pathophysiological events occur over the span of 1 wk of UUO, making this an attractive model for study. This paper reviews some of the ongoing studies that utilized a rodent model of UUO. Some of the findings of the animal model have been compared with observations made in patients with obstructive nephropathy. Most of the evidence suggests that the rodent model of UUO is reflective of human renal disease processes.
Collapse
Affiliation(s)
- Saulo Klahr
- Department of Internal Medicine, Washington University School of Medicine at Barnes-Jewish Hospital, St. Louis, Missouri 63110-1092, USA.
| | | |
Collapse
|