1
|
Cheong A, Craciun F, Husson H, Gans J, Escobedo J, Chang YC, Guo L, Goncalves M, Kaplan N, Smith LA, Moreno S, Boulanger J, Liu S, Saleh J, Zhang M, Blazier AS, Qiu W, Macklin A, Iyyanki T, Chatelain C, Khader S, Natoli TA, Ibraghimov-Beskrovnaya O, Ofengeim D, Proto JD. Glucosylceramide synthase modulation ameliorates murine renal pathologies and promotes macrophage effector function in vitro. Commun Biol 2024; 7:932. [PMID: 39095617 PMCID: PMC11297156 DOI: 10.1038/s42003-024-06606-7] [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: 02/01/2024] [Accepted: 07/19/2024] [Indexed: 08/04/2024] Open
Abstract
While significant advances have been made in understanding renal pathophysiology, less is known about the role of glycosphingolipid (GSL) metabolism in driving organ dysfunction. Here, we used a small molecule inhibitor of glucosylceramide synthase to modulate GSL levels in three mouse models of distinct renal pathologies: Alport syndrome (Col4a3 KO), polycystic kidney disease (Nek8jck), and steroid-resistant nephrotic syndrome (Nphs2 cKO). At the tissue level, we identified a core immune-enriched transcriptional signature that was shared across models and enriched in human polycystic kidney disease. Single nuclei analysis identified robust transcriptional changes across multiple kidney cell types, including epithelial and immune lineages. To further explore the role of GSL modulation in macrophage biology, we performed in vitro studies with homeostatic and inflammatory bone marrow-derived macrophages. Cumulatively, this study provides a comprehensive overview of renal dysfunction and the effect of GSL modulation on kidney-derived cells in the setting of renal dysfunction.
Collapse
Affiliation(s)
- Agnes Cheong
- Rare and Neurologic Diseases Research, Sanofi, Cambridge, MA, USA.
| | | | - Hervé Husson
- Genomics Medicine Unit, Sanofi, Waltham, MA, USA
| | - Joseph Gans
- Translational Sciences, Sanofi, Cambridge, MA, USA
| | | | | | - Lilu Guo
- Translational Sciences, Sanofi, Cambridge, MA, USA
| | | | - Nathan Kaplan
- Rare and Neurologic Diseases Research, Sanofi, Cambridge, MA, USA
| | - Laurie A Smith
- Rare and Neurologic Diseases Research, Sanofi, Cambridge, MA, USA
| | - Sarah Moreno
- Rare and Neurologic Diseases Research, Sanofi, Cambridge, MA, USA
| | - Joseph Boulanger
- Research and Development Business Office, Sanofi, Cambridge, MA, USA
| | - Shiguang Liu
- Rare Diseases and Rare Blood Disorders Research, Sanofi, Cambridge, MA, USA
| | - Jacqueline Saleh
- Rare and Neurologic Diseases Research, Sanofi, Cambridge, MA, USA
| | - Mindy Zhang
- Translational Sciences, Sanofi, Cambridge, MA, USA
| | - Anna S Blazier
- Rare and Neurologic Diseases Research, Sanofi, Cambridge, MA, USA
| | - Weiliang Qiu
- Non-Clinical Efficacy & Safety, Sanofi, Cambridge, MA, USA
| | - Andrew Macklin
- Rare and Neurologic Diseases Research, Sanofi, Cambridge, MA, USA
| | - Tejaswi Iyyanki
- Precision Medicine and Computational Biology, Sanofi, Cambridge, MA, USA
| | - Clément Chatelain
- Precision Medicine and Computational Biology, Sanofi, Cambridge, MA, USA
| | - Shameer Khader
- Precision Medicine and Computational Biology, Sanofi, Cambridge, MA, USA
| | - Thomas A Natoli
- Rare and Neurologic Diseases Research, Sanofi, Cambridge, MA, USA
| | | | - Dimitry Ofengeim
- Rare and Neurologic Diseases Research, Sanofi, Cambridge, MA, USA
| | - Jonathan D Proto
- Rare and Neurologic Diseases Research, Sanofi, Cambridge, MA, USA.
| |
Collapse
|
2
|
Maryam B, Smith ME, Miller SJ, Natarajan H, Zimmerman KA. Macrophage Ontogeny, Phenotype, and Function in Ischemia Reperfusion-Induced Injury and Repair. KIDNEY360 2024; 5:459-470. [PMID: 38297436 PMCID: PMC11000738 DOI: 10.34067/kid.0000000000000376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/22/2024] [Indexed: 02/02/2024]
Abstract
AKI is characterized by a sudden, and usually reversible, decline in kidney function. In mice, ischemia-reperfusion injury (IRI) is commonly used to model the pathophysiologic features of clinical AKI. Macrophages are a unifying feature of IRI as they regulate both the initial injury response as well as the long-term outcome following resolution of injury. Initially, macrophages in the kidney take on a proinflammatory phenotype characterized by the production of inflammatory cytokines, such as CCL2 (monocyte chemoattractant protein 1), IL-6, IL-1 β , and TNF- α . Release of these proinflammatory cytokines leads to tissue damage. After resolution of the initial injury, macrophages take on a reparative role, aiding in tissue repair and restoration of kidney function. By contrast, failure to resolve the initial injury results in prolonged inflammatory macrophage accumulation and increased kidney damage, fibrosis, and the eventual development of CKD. Despite the extensive amount of literature that has ascribed these functions to M1/M2 macrophages, a recent paradigm shift in the macrophage field now defines macrophages on the basis of their ontological origin, namely monocyte-derived and tissue-resident macrophages. In this review, we focus on macrophage phenotype and function during IRI-induced injury, repair, and transition to CKD using both the classic (M1/M2) and novel (ontological origin) definition of kidney macrophages.
Collapse
Affiliation(s)
- Bibi Maryam
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Morgan E. Smith
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Sarah J. Miller
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Hariharasudan Natarajan
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kurt A. Zimmerman
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
3
|
Torrico S, Hotter G, Játiva S. Development of Cell Therapies for Renal Disease and Regenerative Medicine. Int J Mol Sci 2022; 23:ijms232415943. [PMID: 36555585 PMCID: PMC9783572 DOI: 10.3390/ijms232415943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
The incidence of renal disease is gradually increasing worldwide, and this condition has become a major public health problem because it is a trigger for many other chronic diseases. Cell therapies using multipotent mesenchymal stromal cells, hematopoietic stem cells, macrophages, and other cell types have been used to induce regeneration and provide a cure for acute and chronic kidney disease in experimental models. This review describes the advances in cell therapy protocols applied to acute and chronic kidney injuries and the attempts to apply these treatments in a clinical setting.
Collapse
Affiliation(s)
- Selene Torrico
- M2rlab-XCELL, 28010 Madrid, Spain
- Department of Experimental Pathology, Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas Institut d’Investigacions Biomèdiques August Pi i Sunyer (IIBB-CSIC-IDIBAPS), 08036 Barcelona, Spain
- Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Georgina Hotter
- Department of Experimental Pathology, Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas Institut d’Investigacions Biomèdiques August Pi i Sunyer (IIBB-CSIC-IDIBAPS), 08036 Barcelona, Spain
- CIBER-BBN, Networking Center on Bioengineering, Biomaterials and Nanomedicine, 50018 Zaragoza, Spain
- Correspondence: (G.H.); (S.J.)
| | - Soraya Játiva
- M2rlab-XCELL, 28010 Madrid, Spain
- Department of Experimental Pathology, Instituto de Investigaciones Biomédicas de Barcelona-Consejo Superior de Investigaciones Científicas Institut d’Investigacions Biomèdiques August Pi i Sunyer (IIBB-CSIC-IDIBAPS), 08036 Barcelona, Spain
- Correspondence: (G.H.); (S.J.)
| |
Collapse
|
4
|
Smilde BJ, Botman E, de Vries TJ, de Vries R, Micha D, Schoenmaker T, Janssen JJWM, Eekhoff EMW. A Systematic Review of the Evidence of Hematopoietic Stem Cell Differentiation to Fibroblasts. Biomedicines 2022; 10:biomedicines10123063. [PMID: 36551819 PMCID: PMC9775738 DOI: 10.3390/biomedicines10123063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/16/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Fibroblasts have an important role in the maintenance of the extracellular matrix of connective tissues by producing and remodelling extracellular matrix proteins. They are indispensable for physiological processes, and as such also associate with many pathological conditions. In recent years, a number of studies have identified donor-derived fibroblasts in various tissues of bone marrow transplant recipients, while others could not replicate these findings. In this systematic review, we provide an overview of the current literature regarding the differentiation of hematopoietic stem cells into fibroblasts in various tissues. PubMed, Embase, and Web of Science (Core Collection) were systematically searched for original articles concerning fibroblast origin after hematopoietic stem cell transplantation in collaboration with a medical information specialist. Our search found 5421 studies, of which 151 were analysed for full-text analysis by two authors independently, resulting in the inclusion of 104 studies. Only studies in animals and humans, in which at least one marker was used for fibroblast identification, were included. The results were described per organ of fibroblast engraftment. We show that nearly all mouse and human organs show evidence of fibroblasts of hematopoietic stem cell transfer origin. Despite significant heterogeneity in the included studies, most demonstrate a significant presence of fibroblasts of hematopoietic lineage in non-hematopoietic tissues. This presence appears to increase after the occurrence of tissue damage.
Collapse
Affiliation(s)
- Bernard J. Smilde
- Department of Internal Medicine Section Endocrinology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Movement Sciences, 1081 HV Amsterdam, The Netherlands
| | - Esmée Botman
- Department of Internal Medicine Section Endocrinology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Movement Sciences, 1081 HV Amsterdam, The Netherlands
| | - Teun J. de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University, 1081 LA Amsterdam, The Netherlands
| | - Ralph de Vries
- Medical Library, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Dimitra Micha
- Department of Human Genetics, Amsterdam University Medical Center, 1081 HV Amsterdam, The Netherlands
| | - Ton Schoenmaker
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University, 1081 LA Amsterdam, The Netherlands
| | | | - Elisabeth M. W. Eekhoff
- Department of Internal Medicine Section Endocrinology, Amsterdam UMC Location Vrije Universiteit Amsterdam, 1081 HV Amsterdam, The Netherlands
- Amsterdam Movement Sciences, 1081 HV Amsterdam, The Netherlands
- Correspondence: ; Tel.: +31-72-548-4444
| |
Collapse
|
5
|
Wei W, Zhao Y, Zhang Y, Jin H, Shou S. The role of IL-10 in kidney disease. Int Immunopharmacol 2022; 108:108917. [DOI: 10.1016/j.intimp.2022.108917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 05/26/2022] [Accepted: 05/30/2022] [Indexed: 02/07/2023]
|
6
|
Spontaneous Polycystic Kidneys with Chronic Renal Failure in an Aged House Musk Shrew (Suncus murinus). Vet Sci 2022; 9:vetsci9030123. [PMID: 35324851 PMCID: PMC8953240 DOI: 10.3390/vetsci9030123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/01/2022] [Accepted: 03/07/2022] [Indexed: 11/17/2022] Open
Abstract
Polycystic kidney disease is one of the most common inheritable renal diseases, characterized by the formation of multiple fluid-filled renal cysts. This disease is a progressive and unfortunately incurable condition. A case of polycystic kidney with chronic renal failure in house musk shrew (Suncus murinus) is described. At clinical presentation, a 16-month-old Suncus murinus showed weight loss and coarse fur. Regarding the biochemical profile, total protein concentrations increased, resulting in a declined albumin: globulin ratio. Blood urea nitrogen and creatinine concentrations were markedly elevated, indicating the end stage of chronic renal failure. Serum amyloid A levels increased and revealed inflammatory reaction during the cyst formation. Histopathologically, multiple cysts were lined by a single layer of epithelial cells or low cuboidal epithelium. The contents were homogenous eosinophilic materials (mucopolysaccharides or mucoproteins) and these cysts contained abundant macrophages. There were also regeneration and dilatation of renal tubes and interstitial fibrosis. The atrophic glomeruli and glomerular capsules were thickened and hyalinized by dense amorphous mucopolysaccharides. These histopathological findings suggested that the pathogenesis of polycystic kidney disease shared a common mechanistic feature across species.
Collapse
|
7
|
Malone AF. Monocytes and Macrophages in Kidney Transplantation and Insights from Single Cell RNA-Seq Studies. KIDNEY360 2021; 2:1654-1659. [PMID: 35372970 PMCID: PMC8785783 DOI: 10.34067/kid.0003842021] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 08/16/2021] [Indexed: 02/04/2023]
Abstract
Single-cell RNA sequencing (scRNA-seq) is a powerful technology that allows for the identification of minority cell types in complex tissues, such as immune cells in the kidney. Previously, gene expression from infrequent cell types was missed using bulk RNA-sequencing methods due to an averaging effect. Additionally, scRNA-seq facilitates assignment of cell origin in a sample, a shortcoming of previous bulk sequencing technologies. Thus, scRNA-seq is ideal to study the immune cell landscape and the alloimmune response in the human kidney transplant. However, there are few studies published to date. Macrophages are known to play an important role in health and disease in the kidney. Furthermore, it is known that macrophages play key roles in rejection of the kidney transplant. The definition, ontogeny, and function of these cells is complex and nomenclature has evolved as new technologies have become available. In this review, an overview is provided of monocyte and macrophage nomenclature, ontogeny, and function, with a specific focus on kidney transplantation, and including novel scRNA-seq findings. scRNA-seq offers an unbiased transcriptional approach to defining macrophages and provides insights into macrophage ontogeny and function not possible with contemporary methods.
Collapse
Affiliation(s)
- Andrew F. Malone
- Division of Nephrology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri
| |
Collapse
|
8
|
Jang HS, Noh MR, Ha L, Kim J, Padanilam BJ. Proximal tubule cyclophilin D mediates kidney fibrogenesis in obstructive nephropathy. Am J Physiol Renal Physiol 2021; 321:F431-F442. [PMID: 34396791 PMCID: PMC8560409 DOI: 10.1152/ajprenal.00171.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 07/22/2021] [Accepted: 08/02/2021] [Indexed: 12/20/2022] Open
Abstract
The proximal tubule (PT) is highly vulnerable to acute injury, including ischemic insult and nephrotoxins, and chronic kidney injury. It has been established that PT injury is a primary cause of the development of chronic kidney disease, but the underlying molecular mechanism remains to be defined. Here, we tested whether PT cyclophilin D (CypD), a mitochondrial matrix protein, is a critical factor to cause kidney fibrosis progression. To define the role of CypD in kidney fibrosis, we used an established mouse model for kidney fibrosis: the unilateral ureteral obstruction (UUO) model in global and PT-specific CypD knockout (KO). Global CypD KO blunted kidney fibrosis progression with inhibition of myofibroblast activation and fibrosis. UUO-induced tubular atrophy was suppressed in kidneys of global CypD KO but not tubular dilation or apoptotic cell death. PT cell cycle arrest was highly increased in wild-type UUO kidneys but was markedly attenuated in global CypD KO UUO kidneys. The number of macrophages and neutrophils was less in UUO kidneys of global CypD KO than those of wild-type kidneys. Proinflammatory and profibrotic factors were all inhibited in global CypD KO. In line with those of global CypD KO, PT-specific CypD KO also blunted kidney fibrosis progression, along with less tubular atrophy, renal parenchymal loss, cell cycle arrest in PT, and inflammation, indicating a critical role for PT CypD in fibrogenesis. Collectively, our data demonstrate that CypD in the PT is a critical factor contributing to kidney fibrosis in UUO, providing a new paradigm for mitochondria-targeted therapeutics of fibrotic diseases.NEW & NOTEWORTHY It has been established that renal proximal tubule (PT) injury is a primary cause of the development of chronic kidney disease, but the underlying molecular mechanism remains to be defined. Here, we show that cyclophilin D, a mitochondrial matrix protein, in the PT causes kidney fibrogenesis in obstructive nephropathy. Our data suggest that targeting PT cyclophilin D could be beneficial to prevent fibrosis progression.
Collapse
Affiliation(s)
- Hee-Seong Jang
- Department of Urology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Mi Ra Noh
- Department of Urology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Ligyeom Ha
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jinu Kim
- Department of Anatomy, Jeju National University School of Medicine, Jeju, South Korea
- Interdisciplinary Graduate Program in Advanced Convergence Technology & Science, Jeju National University, Jeju, South Korea
| | - Babu J Padanilam
- Department of Urology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
9
|
Jiao B, An C, Tran M, Du H, Wang P, Zhou D, Wang Y. Pharmacological Inhibition of STAT6 Ameliorates Myeloid Fibroblast Activation and Alternative Macrophage Polarization in Renal Fibrosis. Front Immunol 2021; 12:735014. [PMID: 34512669 PMCID: PMC8426438 DOI: 10.3389/fimmu.2021.735014] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/04/2021] [Indexed: 12/24/2022] Open
Abstract
A hallmark of chronic kidney disease is renal fibrosis, which can result in progressive loss of kidney function. Currently, there is no effective therapy for renal fibrosis. Therefore, there is an urgent need to identify potential drug targets for renal fibrosis. In this study, we examined the effect of a selective STAT6 inhibitor, AS1517499, on myeloid fibroblast activation, macrophage polarization, and development of renal fibrosis in two experimental murine models. To investigate the effect of STAT6 inhibition on myeloid fibroblast activation, macrophage polarization, and kidney fibrosis, wild-type mice were subjected to unilateral ureteral obstruction or folic acid administration and treated with AS1517499. Mice treated with vehicle were used as control. At the end of experiments, kidneys were harvested for analysis of myeloid fibroblast activation, macrophage polarization, and renal fibrosis and function. Unilateral ureteral obstruction or folic acid administration induced STAT6 activation in interstitial cells of the kidney, which was significantly abolished by AS1517499 treatment. Mice treated with AS1517499 accumulated fewer myeloid fibroblasts and myofibroblasts in the kidney with ureteral obstruction or folic acid nephropathy compared with vehicle-treated mice. Moreover, AS1517499 significantly suppressed M2 macrophage polarization in the injured kidney. Furthermore, AS1517499 markedly reduced the expression levels of extracellular matrix proteins, and development of kidney fibrosis and dysfunction. These findings suggest that AS1517499 inhibits STAT6 activation, suppresses myeloid fibroblast activation, reduces M2 macrophage polarization, attenuates extracellular matrix protein production, and preserves kidney function. Therefore, targeting STAT6 with AS1517499 is a novel therapeutic approach for chronic kidney disease.
Collapse
Affiliation(s)
- Baihai Jiao
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Changlong An
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Melanie Tran
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Hao Du
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Penghua Wang
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Dong Zhou
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Yanlin Wang
- Division of Nephrology, Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, United States
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT, United States
- Institute for Systems Genomics, University of Connecticut School of Medicine, Farmington, CT, United States
- Renal Section, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
10
|
Abstract
PURPOSE OF REVIEW Macrophages play an important role in regulating homeostasis, kidney injury, repair, and tissue fibrogenesis. The present review will discuss recent advances that explore the novel subsets and functions of macrophage in the pathogenesis of kidney damage and hypertension. RECENT FINDINGS Macrophages differentiate into a variety of subsets in microenvironment-dependent manner. Although the M1/M2 nomenclature is still applied in considering the pro-inflammatory versus anti-inflammatory effects of macrophages in kidney injury, novel, and accurate macrophage phenotypes are defined by flow cytometric markers and single-cell RNA signatures. Studies exploring the crosstalk between macrophages and other cells are rapidly advancing with the additional recognition of exosome trafficking between cells. Using murine conditional mutants, actions of macrophage can be defined more precisely than in bone marrow transfer models. Some studies revealed the opposing effects of the same protein in renal parenchymal cells and macrophages, highlighting a need for the development of cell-specific immune therapies for translation. SUMMARY Macrophage-targeted therapies hold potential for limiting kidney injury and hypertension. To realize this potential, future studies will be required to understand precise mechanisms in macrophage polarization, crosstalk, proliferation, and maturation in the setting of renal disease.
Collapse
|
11
|
Abstract
Interstitial inflammation is an important feature of cystic kidney disease. Renal macrophages are the most well-studied inflammatory cell in the kidney, and their involvement in cyst formation has been reported in different animal models and patients with cystic kidney disease. Originally, it was believed that renal macrophages were maintained from a constant supply of bone marrow-derived circulating monocytes, and could be recruited to the kidney in response to local inflammation. However, this idea has been challenged using fate-mapping methods, by showing that at least two distinct developmental origins of macrophages are present in the adult mouse kidney. The first type, infiltrating macrophages, are recruited from circulating monocytes and gradually develop macrophage properties on entering the kidney. The second, resident macrophages, predominantly originate from embryonic precursors, colonize the kidney during its development, and proliferate in situ to maintain their population throughout adulthood. Infiltrating and resident macrophages work together to maintain homeostasis and properly respond to pathologic conditions, such as AKI, cystic kidney disease, or infection. This review will briefly summarize current knowledge of resident macrophages in cystic kidney disease.
Collapse
Affiliation(s)
- Zhang Li
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kurt A. Zimmerman
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Bradley K. Yoder
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
12
|
Lantz C, Radmanesh B, Liu E, Thorp EB, Lin J. Single-cell RNA sequencing uncovers heterogenous transcriptional signatures in macrophages during efferocytosis. Sci Rep 2020; 10:14333. [PMID: 32868786 PMCID: PMC7459098 DOI: 10.1038/s41598-020-70353-y] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 07/23/2020] [Indexed: 12/24/2022] Open
Abstract
Efferocytosis triggers cellular reprogramming, including the induction of mRNA transcripts which encode anti-inflammatory cytokines that promote inflammation resolution. Our current understanding of this transcriptional response is largely informed from analysis of bulk phagocyte populations; however, this precludes the resolution of heterogeneity between individual macrophages and macrophage subsets. Moreover, phagocytes may contain so called "passenger" transcripts that originate from engulfed apoptotic bodies, thus obscuring the true transcriptional reprogramming of the phagocyte. To define the transcriptional diversity during efferocytosis, we utilized single-cell mRNA sequencing after co-cultivating macrophages with apoptotic cells. Importantly, transcriptomic analyses were performed after validating the disappearance of apoptotic cell-derived RNA sequences. Our findings reveal new heterogeneity of the efferocytic response at a single-cell resolution, particularly evident between F4/80+ MHCIILO and F4/80- MHCIIHI macrophage sub-populations. After exposure to apoptotic cells, the F4/80+ MHCIILO subset significantly induced pathways associated with tissue and cellular homeostasis, while the F4/80- MHCIIHI subset downregulated these putative signaling axes. Ablation of a canonical efferocytosis receptor, MerTK, blunted efferocytic signatures and led to the escalation of cell death-associated transcriptional signatures in F4/80+ MHCIILO macrophages. Taken together, our results newly elucidate the heterogenous transcriptional response of single-cell peritoneal macrophages after exposure to apoptotic cells.
Collapse
Affiliation(s)
- Connor Lantz
- Department of Pathology, Pediatrics, Medicine and the Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Evanston, USA.
| | - Behram Radmanesh
- Department of Pathology, Pediatrics, Medicine and the Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Evanston, USA
| | - Esther Liu
- Department of Pathology, Pediatrics, Medicine and the Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Evanston, USA
| | - Edward B Thorp
- Department of Pathology, Pediatrics, Medicine and the Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Evanston, USA
| | - Jennie Lin
- Department of Pathology, Pediatrics, Medicine and the Feinberg Cardiovascular and Renal Research Institute, Feinberg School of Medicine, Northwestern University, Evanston, USA
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL, 60611, USA
| |
Collapse
|
13
|
Macrophages: versatile players in renal inflammation and fibrosis. Nat Rev Nephrol 2019; 15:144-158. [PMID: 30692665 DOI: 10.1038/s41581-019-0110-2] [Citation(s) in RCA: 595] [Impact Index Per Article: 99.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2018] [Indexed: 12/15/2022]
Abstract
Macrophages have important roles in immune surveillance and in the maintenance of kidney homeostasis; their response to renal injury varies enormously depending on the nature and duration of the insult. Macrophages can adopt a variety of phenotypes: at one extreme, M1 pro-inflammatory cells contribute to infection clearance but can also promote renal injury; at the other extreme, M2 anti-inflammatory cells have a reparative phenotype and can contribute to the resolution phase of the response to injury. In addition, bone marrow monocytes can differentiate into myeloid-derived suppressor cells that can regulate T cell immunity in the kidney. However, macrophages can also promote renal fibrosis, a major driver of progression to end-stage renal disease, and the CD206+ subset of M2 macrophages is strongly associated with renal fibrosis in both human and experimental diseases. Myofibroblasts are important contributors to renal fibrosis and recent studies provide evidence that macrophages recruited from the bone marrow can transition directly into myofibroblasts within the injured kidney. This process is termed macrophage-to-myofibroblast transition (MMT) and is driven by transforming growth factor-β1 (TGFβ1)-Smad3 signalling via a Src-centric regulatory network. MMT may serve as a key checkpoint for the progression of chronic inflammation into pathogenic fibrosis.
Collapse
|
14
|
Sun J, Wu Y, Long C, He P, Gu J, Yang L, Liang Y, Wang Y. Anthocyanins isolated from blueberry ameliorates CCl4 induced liver fibrosis by modulation of oxidative stress, inflammation and stellate cell activation in mice. Food Chem Toxicol 2018; 120:491-499. [DOI: 10.1016/j.fct.2018.07.048] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/15/2018] [Accepted: 07/26/2018] [Indexed: 02/07/2023]
|
15
|
Williams TM, Wise AF, Layton DS, Ricardo SD. Phenotype and influx kinetics of leukocytes and inflammatory cytokine production in kidney ischemia/reperfusion injury. Nephrology (Carlton) 2018; 23:75-85. [PMID: 27696567 DOI: 10.1111/nep.12941] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Revised: 09/26/2016] [Accepted: 09/29/2016] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND AIM Kidney ischemia/reperfusion (IR) injury is characterized by tubular epithelial cell (TEC) death and an inflammatory response involving cytokine production and immune cell infiltration. In various kidney diseases, increased macrophage numbers correlate with injury severity and poor prognosis. However, macrophage plasticity enables a diverse range of functions, including wound healing, making them a key target for novel therapies. This study aimed to comprehensively characterize the changes in myeloid and epithelial cells and the production of cytokines throughout the experimental IR model of acute kidney injury to aid in the identification of targets to promote and enhance kidney regeneration and repair. METHODS Flow cytometric analysis of murine unilateral IR injury was used to assess TEC and myeloid cell subpopulations in conjunction with histological analysis and cytokine production at 6 h, 1, 3, 5 and 7 days post IR injury, spanning the initial inflammatory phase and the following reparative phase. RESULTS IR injury resulted in a rapid infiltration of Ly6Chigh monocytes and neutrophils with a steady rise in F4/80high MHCIIhigh macrophages over the injury time. The production of the inflammatory cytokines IL-6, MCP-1 and TNF coincided with an increase in IL-10 production. CONCLUSION This characterization will provide a reference point for future studies designed to manipulate immune cell phenotype and function in order to promote endogenous repair of damaged kidneys.
Collapse
Affiliation(s)
- Timothy M Williams
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Andrea F Wise
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| | - Daniel S Layton
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia.,Biosecurity Flagship, CSIRO, Geelong, Australia
| | - Sharon D Ricardo
- Department of Anatomy and Developmental Biology, Monash University, Melbourne, Australia
| |
Collapse
|
16
|
Park JH, Ku HJ, Kim JK, Park JW, Lee JH. Amelioration of High Fructose-Induced Cardiac Hypertrophy by Naringin. Sci Rep 2018; 8:9464. [PMID: 29930336 PMCID: PMC6013481 DOI: 10.1038/s41598-018-27788-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/31/2018] [Indexed: 12/12/2022] Open
Abstract
Heart failure is a frequent unfavorable outcome of pathological cardiac hypertrophy. Recent increase in dietary fructose consumption mirrors the rise in prevalence of cardiovascular diseases such as cardiac hypertrophy leading to concerns raised by public health experts. Mitochondria, comprising 30% of cardiomyocyte volume, play a central role in modulating redox-dependent cellular processes such as metabolism and apoptosis. Furthermore, mitochondrial dysfunction is a key cause of pathogenesis of fructose-induced cardiac hypertrophy. Naringin, a major flavanone glycoside in citrus species, has displayed strong antioxidant potential in models of oxidative stress. In this study, we evaluated protective effects of naringin against fructose-induced cardiac hypertrophy and associated mechanisms of action, using in vitro and in vivo models. We found that naringin suppressed mitochondrial ROS production and mitochondrial dysfunction in cardiomyocytes exposed to fructose and consequently reduced cardiomyocyte hypertrophy by regulating AMPK-mTOR signaling axis. Furthermore, naringin counteracted fructose-induced cardiomyocyte apoptosis, and this function of naringin was linked to its ability to inhibit ROS-dependent ATM-mediated p53 signaling. This result was supported by observations in in vivo mouse model of cardiac hypertrophy. These findings indicate a novel role for naringin in protecting against fructose-induced cardiac hypertrophy and suggest unique therapeutic strategies for prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Jung Hyun Park
- Department of Food and Biotechnology, Korea University, Sejong, Korea
| | - Hyeong Jun Ku
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Taegu, Korea
| | - Jae Kyeom Kim
- School of Human Environmental Sciences, University of Arkansas, Fayetteville, Arkansas, USA
| | - Jeen-Woo Park
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Taegu, Korea.
| | - Jin Hyup Lee
- Department of Food and Biotechnology, Korea University, Sejong, Korea.
| |
Collapse
|
17
|
Park JH, Ku HJ, Lee JH, Park JW. IDH2 deficiency accelerates skin pigmentation in mice via enhancing melanogenesis. Redox Biol 2018; 17:16-24. [PMID: 29660504 PMCID: PMC6006679 DOI: 10.1016/j.redox.2018.04.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/03/2018] [Accepted: 04/05/2018] [Indexed: 01/18/2023] Open
Abstract
Melanogenesis is a complex biosynthetic pathway regulated by multiple agents, which are involved in the production, transport, and release of melanin. Melanin has diverse roles, including determination of visible skin color and photoprotection. Studies indicate that melanin synthesis is tightly linked to the interaction between melanocytes and keratinocytes. α-melanocyte-stimulating hormone (α-MSH) is known as a trigger that enhances melanin biosynthesis in melanocytes through paracrine effects. Accumulated reactive oxygen species (ROS) in skin affects both keratinocytes and melanocytes by causing DNA damage, which eventually leads to the stimulation of α-MSH production. Mitochondria are one of the main sources of ROS in the skin and play a central role in modulating redox-dependent cellular processes such as metabolism and apoptosis. Therefore, mitochondrial dysfunction may serve as a key for the pathogenesis of skin melanogenesis. Mitochondrial NADP+-dependent isocitrate dehydrogenase (IDH2) is a key enzyme that regulates mitochondrial redox balance and reduces oxidative stress-induced cell injury through the generation of NADPH. Downregulation of IDH2 expression resulted in an increase in oxidative DNA damage in mice skin through ROS-dependent ATM-mediated p53 signaling. IDH2 deficiency also promoted pigmentation on the dorsal skin of mice, as evident from the elevated levels of melanin synthesis markers. Furthermore, pretreatment with mitochondria-targeted antioxidant mito-TEMPO alleviated oxidative DNA damage and melanogenesis induced by IDH2 deficiency both in vitro and in vivo. Together, our findings highlight the role of IDH2 in skin melanogenesis in association with mitochondrial ROS and suggest unique therapeutic strategies for the prevention of skin pigmentation. Melanogenesis is associated with the production of ROS. IDH2 is an essential enzyme in the mitochondrial antioxidant system. Downregulation of IDH2 induces ROS-dependent ATM-mediated p53 signaling. IDH2 deficiency promotes skin pigmentation. mito-TEMPO alleviates melanogenesis caused by IDH2 deficiency.
Collapse
Affiliation(s)
- Jung Hyun Park
- Department of Food and Biotechnology, Korea University, Sejong, Republic of Korea
| | - Hyeong Jun Ku
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Taegu, Republic of Korea
| | - Jin Hyup Lee
- Department of Food and Biotechnology, Korea University, Sejong, Republic of Korea.
| | - Jeen-Woo Park
- School of Life Sciences and Biotechnology, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Taegu, Republic of Korea.
| |
Collapse
|
18
|
Ghosh M, Thangada S, Dasgupta O, Khanna KM, Yamase HT, Kashgarian M, Hla T, Shapiro LH, Ferrer FA. Cell-intrinsic sphingosine kinase 2 promotes macrophage polarization and renal inflammation in response to unilateral ureteral obstruction. PLoS One 2018. [PMID: 29518138 PMCID: PMC5843290 DOI: 10.1371/journal.pone.0194053] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Sphingosine Kinase-2 (Sphk2) is responsible for the production of the bioactive lipid Sphingosine-1 Phosphate, a key regulator of tissue repair. Here we address the in vivo significance of Sphingosine Kinase -2 in renal inflammation/fibrosis in response to unilateral ureteral obstruction using both genetic and pharmacological strategies. Obstructed kidneys of Sphk2-/- mice showed reduced renal damage and diminished levels of the renal injury markers TGFβ1 and αSMA when compared to wild type controls. We found a consistently significant increase in anti-inflammatory (M2) macrophages in obstructed Sphk2-/- kidneys by flow cytometry and a decrease in mRNA levels of the inflammatory cytokines, MCP1, TNFα, CXCL1 and ILβ1, suggesting an anti-inflammatory bias in the absence of Sphk2. Indeed, metabolic profiling showed that the pro-inflammatory glycolytic pathway is largely inactive in Sphk2-/- bone marrow-derived macrophages. Furthermore, treatment with the M2-promoting cytokines IL-4 or IL-13 demonstrated that macrophages lacking Sphk2 polarized more efficiently to the M2 phenotype than wild type cells. Bone marrow transplant studies indicated that expression of Sphk2-/- on either the hematopoietic or parenchymal cells did not fully rescue the pro-healing phenotype, confirming that both infiltrating M2-macrophages and the kidney microenvironment contribute to the damaging Sphk2 effects. Importantly, obstructed kidneys from mice treated with an Sphk2 inhibitor recapitulated findings in the genetic model. These results demonstrate that reducing Sphk2 activity by genetic or pharmacological manipulation markedly decreases inflammatory and fibrotic responses to obstruction, resulting in diminished renal injury and supporting Sphk2 as a novel driver of the pro-inflammatory macrophage phenotype.
Collapse
Affiliation(s)
- Mallika Ghosh
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, United States of America
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT, United States of America
| | - Shobha Thangada
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, United States of America
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT, United States of America
| | - Oisharya Dasgupta
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, United States of America
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT, United States of America
| | - Kamal M. Khanna
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States of America
| | - Harold T. Yamase
- Department of Pathology, University of Connecticut School of Medicine, Farmington, CT, United States of America
| | - Michael Kashgarian
- Department of Pathology, Yale University Cancer Research Center, New Haven, CT, United States of America
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, United States of America
| | - Linda H. Shapiro
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, United States of America
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT, United States of America
- * E-mail: (FAF); (LHS)
| | - Fernando A. Ferrer
- Center for Vascular Biology, University of Connecticut School of Medicine, Farmington, CT, United States of America
- Department of Cell Biology, University of Connecticut School of Medicine, Farmington, CT, United States of America
- Section of Pediatric Urology, Children's Hospital of Omaha, Department of Surgery, University of Nebraska School of Medicine, Omaha, NE, United States of America
- * E-mail: (FAF); (LHS)
| |
Collapse
|
19
|
Munro DAD, Hughes J. The Origins and Functions of Tissue-Resident Macrophages in Kidney Development. Front Physiol 2017; 8:837. [PMID: 29118719 PMCID: PMC5660965 DOI: 10.3389/fphys.2017.00837] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 10/09/2017] [Indexed: 12/21/2022] Open
Abstract
The adult kidney hosts tissue-resident macrophages that can cause, prevent, and/or repair renal damage. Most of these macrophages derive from embryonic progenitors that colonize the kidney during its development and proliferate in situ throughout adulthood. Although the precise origins of kidney macrophages remain controversial, recent studies have revealed that embryonic macrophage progenitors initially migrate from the yolk sac, and later from the fetal liver, into the developing kidney. Once in the kidney, tissue-specific transcriptional regulators specify macrophage progenitors into dedicated kidney macrophages. Studies suggest that kidney macrophages facilitate many processes during renal organogenesis, such as branching morphogenesis and the clearance of cellular debris; however, little is known about how the origins and specification of kidney macrophages dictate their function. Here, we review significant new findings about the origins, specification, and developmental functions of kidney macrophages.
Collapse
Affiliation(s)
- David A D Munro
- Centre for Integrative Physiology, School of Biomedical Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Jeremy Hughes
- MRC Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
20
|
Pushpakumar S, Ren L, Kundu S, Gamon A, Tyagi SC, Sen U. Toll-like Receptor 4 Deficiency Reduces Oxidative Stress and Macrophage Mediated Inflammation in Hypertensive Kidney. Sci Rep 2017; 7:6349. [PMID: 28743964 PMCID: PMC5526876 DOI: 10.1038/s41598-017-06484-6] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 06/13/2017] [Indexed: 12/22/2022] Open
Abstract
Oxidative stress and inflammation are integral to hypertension-induced renal injury. A unifying feature for the two components is Toll-like receptors (TLR), which are key regulators of the innate immune system. Recent studies implicate TLR4 activation and oxidative stress in cardiovascular diseases and also as a link between inflammation and hypertension. However, its role in hypertension induced renal injury remains unexplored. In the present study, we investigated whether TLR-4 deficiency reduces Ang-II-induced renal injury and fibrosis by attenuating reactive oxygen species (ROS) production and inflammation. C3H/HeOuJ mice with normal TLR-4 and C3H/HeJ Lps-d with dysfunctional TLR4 (TLR4 deficiency) were treated without or with Ang-II. In response to Ang-II, TLR4 deficient mice had reduced renal resistive index and increased renal cortical blood flow compared to mice with normal TLR4. Further, TLR4 deficiency reduced oxidative stress and increased antioxidant capacity (MnSOD, CuSOD and Catalase activity). TLR4 deficiency was also associated with reduced inflammation (MCP-1, MIP-2, TNF-α, IL-6 and CD68), decreased accumulation of bone marrow-derived fibroblasts and TGF-β expression. Our data suggests that in C3H/HeJ Lps-d mice, deficiency of functional TLR4 reduces oxidative stress and macrophage activation to decrease TGF-β-induced extracellular matrix protein deposition in the kidney in Ang-II induced hypertension.
Collapse
Affiliation(s)
- Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY-40202, USA
| | - Lu Ren
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY-40202, USA
| | - Sourav Kundu
- Institute of Advanced Study in Science and Technology, Guwahati, Assam, 781035, India
| | | | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY-40202, USA
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY-40202, USA.
| |
Collapse
|
21
|
Schnaper HW. The Tubulointerstitial Pathophysiology of Progressive Kidney Disease. Adv Chronic Kidney Dis 2017; 24:107-116. [PMID: 28284376 PMCID: PMC5351778 DOI: 10.1053/j.ackd.2016.11.011] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2016] [Revised: 11/07/2016] [Accepted: 11/13/2016] [Indexed: 02/07/2023]
Abstract
Accumulating evidence suggests that the central locus for the progression of CKD is the renal proximal tubule. As injured tubular epithelial cells dedifferentiate in attempted repair, they stimulate inflammation and recruit myofibroblasts. At the same time, tissue loss stimulates remnant nephron hypertrophy. Increased tubular transport workload eventually exceeds the energy-generating capacity of the hypertrophied nephrons, leading to anerobic metabolism, acidosis, hypoxia, endoplasmic reticulum stress, and the induction of additional inflammatory and fibrogenic responses. The result is a vicious cycle of injury, misdirected repair, maladaptive responses, and more nephron loss. Therapy that might be advantageous at one phase of this progression pathway could be deleterious during other phases. Thus, interrupting this downward spiral requires narrowly targeted approaches that promote healing and adequate function without generating further entry into the progression cycle.
Collapse
Affiliation(s)
- H William Schnaper
- Division of Kidney Diseases, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL.
| |
Collapse
|
22
|
Wang S, Meng XM, Ng YY, Ma FY, Zhou S, Zhang Y, Yang C, Huang XR, Xiao J, Wang YY, Ka SM, Tang YJ, Chung ACK, To KF, Nikolic-Paterson DJ, Lan HY. TGF-β/Smad3 signalling regulates the transition of bone marrow-derived macrophages into myofibroblasts during tissue fibrosis. Oncotarget 2017; 7:8809-22. [PMID: 26684242 PMCID: PMC4891006 DOI: 10.18632/oncotarget.6604] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 11/25/2015] [Indexed: 11/25/2022] Open
Abstract
Myofibroblasts are a main cell-type of collagen-producing cells during tissue fibrosis, but their origins remains controversial. While bone marrow-derived myofibroblasts in renal fibrosis has been reported, the cell origin and mechanisms regulating their transition into myofibroblasts remain undefined. In the present study, cell lineage tracing studies by adoptive transfer of GFP+ or dye-labelled macrophages identified that monocyte/macrophages from bone marrow can give rise to myofibroblasts via the process of macrophage-myofibroblast transition (MMT) in a mouse model of unilateral ureteric obstruction. The MMT cells were a major source of collagen-producing fibroblasts in the fibrosing kidney, accounting for more than 60% of α-SMA+ myofibroblasts. The MMT process occurred predominantly within M2-type macrophages and was regulated by TGF-β/Smad3 signalling as deletion of Smad3 in the bone marrow compartment of GFP+ chimeric mice prevented the M2 macrophage transition into the MMT cells and progressive renal fibrosis. In vitro studies in Smad3 null bone marrow macrophages also showed that Smad3 was required for TGF-β1-induced MMT and collagen production. In conclusion, we have demonstrated that bone marrow-derived fibroblasts originate from the monocyte/macrophage population via a process of MMT. This process contributes to progressive renal tissue fibrosis and is regulated by TGF-β/Smad3 signalling.
Collapse
Affiliation(s)
- Shuang Wang
- Li Ka Shing Institute of Health Sciences, Departments of Medicine and Therapeutics, Chemical Pathology, and Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiao-Ming Meng
- Li Ka Shing Institute of Health Sciences, Departments of Medicine and Therapeutics, Chemical Pathology, and Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yee-Yung Ng
- Division of Nephrology, Department of Medicine, Institute of Clinical Medicine, Taipei Veterans General Hospital, National Yang Ming University, Taipei, Taiwan
| | - Frank Y Ma
- Department of Nephrology and Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Shuang Zhou
- Li Ka Shing Institute of Health Sciences, Departments of Medicine and Therapeutics, Chemical Pathology, and Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yang Zhang
- Li Ka Shing Institute of Health Sciences, Departments of Medicine and Therapeutics, Chemical Pathology, and Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chen Yang
- Li Ka Shing Institute of Health Sciences, Departments of Medicine and Therapeutics, Chemical Pathology, and Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiao-Ru Huang
- Li Ka Shing Institute of Health Sciences, Departments of Medicine and Therapeutics, Chemical Pathology, and Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jun Xiao
- Li Ka Shing Institute of Health Sciences, Departments of Medicine and Therapeutics, Chemical Pathology, and Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ying-Ying Wang
- Li Ka Shing Institute of Health Sciences, Departments of Medicine and Therapeutics, Chemical Pathology, and Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Shuk-Man Ka
- Li Ka Shing Institute of Health Sciences, Departments of Medicine and Therapeutics, Chemical Pathology, and Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yong-Jiang Tang
- Li Ka Shing Institute of Health Sciences, Departments of Medicine and Therapeutics, Chemical Pathology, and Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Arthur C K Chung
- Li Ka Shing Institute of Health Sciences, Departments of Medicine and Therapeutics, Chemical Pathology, and Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ka-Fai To
- Li Ka Shing Institute of Health Sciences, Departments of Medicine and Therapeutics, Chemical Pathology, and Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - David J Nikolic-Paterson
- Department of Nephrology and Monash University Department of Medicine, Monash Medical Centre, Clayton, Victoria, Australia
| | - Hui-Yao Lan
- Li Ka Shing Institute of Health Sciences, Departments of Medicine and Therapeutics, Chemical Pathology, and Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
23
|
Inhibition of microtubule dynamics impedes repair of kidney ischemia/reperfusion injury and increases fibrosis. Sci Rep 2016; 6:27775. [PMID: 27270990 PMCID: PMC4897697 DOI: 10.1038/srep27775] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 05/23/2016] [Indexed: 11/08/2022] Open
Abstract
The microtubule cytoskeleton is composed of α-tubulin and β-tubulin heterodimers, and it serves to regulate the shape, motility, and division of a cell. Post-translational modifications including acetylation are closely associated with the functional aspects of the microtubule, involving in a number of pathological diseases. However, the role of microtubule acetylation in acute kidney injury (AKI) and progression of AKI to chronic kidney disease have yet to be understood. In this study, ischemia/reperfusion (I/R), a major cause of AKI, resulted in deacetylation of the microtubules with a decrease in α-tubulin acetyltransferase 1 (α-TAT1). Paclitaxel (taxol), an agent that stabilizes microtubules by tubulin acetylation, treatment during the recovery phase following I/R injury inhibited tubular cell proliferation, impaired renal functional recovery, and worsened fibrosis. Taxol induced α-tubulin acetylation and post-I/R cell cycle arrest. Taxol aggregated the microtubule in the cytoplasm, resulting in suppression of microtubule dynamics. Our studies have demonstrated for the first time that I/R induced deacetylation of the microtubules, and that inhibition of microtubule dynamics retarded repair of injured tubular epithelial cells leading to an acceleration of fibrosis. This suggests that microtubule dynamics plays an important role in the processes of repair and fibrosis after AKI.
Collapse
|
24
|
Kim JI, Noh MR, Kim KY, Jang HS, Kim HY, Park KM. Methionine sulfoxide reductase A deficiency exacerbates progression of kidney fibrosis induced by unilateral ureteral obstruction. Free Radic Biol Med 2015. [PMID: 26210777 DOI: 10.1016/j.freeradbiomed.2015.07.018] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Methionine sulfoxide reductase A (MsrA), which stereospecifically catalyzes the reduction of methionine-S-sulfoxide, is an important reactive oxygen species (ROS) scavenger. Tissue fibrosis is a maladaptive repair process following injury, associated with oxidative stress. In this study, we investigated the role of MsrA in unilateral ureteral obstruction (UUO)-induced kidney fibrosis and its underlying mechanisms by using MsrA gene-deleted mice (MsrA(-/-)). MsrA deletion increased collagen deposition in the interstitium and the expression of collagen III and α-smooth muscle actin in the UUO kidneys, indicating that MsrA deficiency exacerbated the progression of UUO-induced kidney fibrosis. UUO reduced the kidney expression of MsrA, MsrB1, and MsrB2, thereby decreasing MsrA and MsrB activity. UUO increased hydrogen peroxide and lipid peroxidation levels and the ratio of oxidized glutathione (GSSG) to total glutathione (GSH) in the kidneys. The UUO-induced elevations in the levels of these oxidative stress markers and leukocyte markers were much higher in the MsrA(-/-) than in the MsrA(+/+) kidneys, the latter suggesting that the exacerbated kidney fibrosis in MsrA(-/-) mice was associated with enhanced inflammatory responses. Collectively, our data suggest that MsrA plays a protective role in the progression of UUO-induced kidney fibrosis via suppression of fibrotic responses caused by oxidative stress and inflammation.
Collapse
Affiliation(s)
- Jee In Kim
- Department of Molecular Medicine and MRC, Keimyung University School of Medicine, Daegu 705-717, Republic of Korea
| | - Mi Ra Noh
- Department of Anatomy and BK21 Plus Program, Kyungpook National University School of Medicine, Daegu 700-422, Republic of Korea
| | - Ki Young Kim
- Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine, Daegu 705-717, Republic of Korea
| | - Hee-Seong Jang
- Department of Anatomy and BK21 Plus Program, Kyungpook National University School of Medicine, Daegu 700-422, Republic of Korea
| | - Hwa-Young Kim
- Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine, Daegu 705-717, Republic of Korea.
| | - Kwon Moo Park
- Department of Anatomy and BK21 Plus Program, Kyungpook National University School of Medicine, Daegu 700-422, Republic of Korea.
| |
Collapse
|
25
|
Karim AS, Reese SR, Wilson NA, Jacobson LM, Zhong W, Djamali A. Nox2 is a mediator of ischemia reperfusion injury. Am J Transplant 2015; 15:2888-99. [PMID: 26104383 PMCID: PMC4636908 DOI: 10.1111/ajt.13368] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 04/23/2015] [Accepted: 04/23/2015] [Indexed: 01/25/2023]
Abstract
Delayed graft function (DGF) results from ischemia-reperfusion injury (IRI) and the generation of reactive oxygen species. We hypothesized that NADPH oxidase 2 (Nox2) plays an important role in pathways leading to DGF. We tested this hypothesis in vitro, in an animal model of IRI using wild type and Nox2(-/-) mice, and in patients with DGF. Under hypoxic conditions, primary tubular epithelial cells from Nox2(-/-) mice had reduced expression of MMP2, vimentin, and HSP27. BUN and creatinine levels were significantly increased in both Nox2(-/-) and WT mice at 4 weeks and 6 months after IRI, suggesting the development of acute and chronic kidney injury. At 4 weeks, kidney fibrosis (α-SMA, picrosirius) and oxidative stress (dihydroethidine, HNE) were significantly reduced in Nox2(-/-) mice, confirming the oxidative and pro-fibrotic effects of Nox2. The molecular signature of IRI using genomic analyses demonstrated a significant decline in hypoxia reponse, oxidative stress, fibrosis, and inflammation in Nox2(-/-) mice. Immunohistochemical analyses of pre-implanatation kidney allograft biopsies from patients with subsequent DGF showed significantly greater Nox2 levels and vascular injury compared with patients without DGF. These studies demonstrate that Nox2 is a modulator of IRI and its absence is associated with reduced inflammation, OS, and fibrosis.
Collapse
Affiliation(s)
- Aos S. Karim
- University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Shannon R. Reese
- University of Wisconsin Department of Medicine
- Division of Nephrology, Madison, WI
| | - Nancy A. Wilson
- University of Wisconsin Department of Medicine
- Division of Nephrology, Madison, WI
| | - Lynn M. Jacobson
- University of Wisconsin Department of Medicine
- Division of Nephrology, Madison, WI
| | - Weixiong Zhong
- University of Wisconsin Department of Pathology and Laboratory Medicine
| | - Arjang Djamali
- University of Wisconsin School of Medicine and Public Health, Madison, WI,University of Wisconsin Department of Medicine
- Division of Nephrology, Madison, WI
| |
Collapse
|
26
|
Baba I, Egi Y, Utsumi H, Kakimoto T, Suzuki K. Inhibitory effects of fasudil on renal interstitial fibrosis induced by unilateral ureteral obstruction. Mol Med Rep 2015; 12:8010-20. [PMID: 26498136 PMCID: PMC4758322 DOI: 10.3892/mmr.2015.4467] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 08/25/2015] [Indexed: 02/07/2023] Open
Abstract
Renal fibrosis is the major cause of chronic kidney disease, and the Rho/Rho-associated coiled-coil kinase (ROCK) signaling cascade is involved in the renal fibrotic processes. Several studies have reported that ROCK inhibitors attenuate renal fibrosis. However, the mechanism of this process remains to be fully elucidated. The present study assessed the inhibitory effect of fasudil, a ROCK inhibitor using immunohistochemistry, reverse transcription-quantitative polymerase chain reaction and western blot analyses, in vivo and in vitro, to elucidate the mechanisms underlying renal interstitial fibrosis. In mice induced with unilateral ureteral obstruction (UUO), collagen accumulation, the expression of fibrosis-associated genes and the content of hydroxyproline in the kidney increased 3, 7, and 14 days following UUO. Fasudil attenuated the histological changes, and the production of collagen and extracellular matrix in the UUO kidney. The expression of α-smooth muscle actin (α-SMA) and the transforming growth factor-β (TGFβ)-Smad signaling pathway, and macrophage infiltration were suppressed by fasudil in the kidneys of the UUO mice. The present study also evaluated the role of intrinsic renal cells and infiltrated macrophages using NRK-52E, NRK-49F and RAW264.7 cells. The mRNA and protein expression levels of collagen I and α-SMA increased in the NRK-52E and NRK-49F cells stimulated by TGF-β1. Hydroxyfasudil, a bioactive metabolite of fasudil, attenuated the increase in the mRNA and protein expression levles of α-SMA in the two cell types. However, the reduction in the mRNA expression of collagen I was observed in the NRK-49F cells only. Hydroxyfasudil decreased the mRNA expression of monocyte chemoattractant protein-1 (MCP-1) induced by TGF-β1 in the NRK-52E cells, but not in the NRK-49F cells. In the RAW264.7 cells, the mRNA expression levels of MCP-1, interleukin (IL)-1β, IL-6 and tumor necrosis factor α were increased significantly following lipopolysaccharide stimulation, and were not suppressed by hydroxyfasudil. These data suggested that the inhibition of ROCK activity by fasudil suppressed the transformation of renal intrinsic cells into the myofibroblast cells, and attenuated the infiltration of macrophages, without inhibiting the expression or the activation of cytokine/chemokines, in the progression of renal interstitial fibrosis.
Collapse
Affiliation(s)
- Itsuko Baba
- Pharmacology Research Laboratories II, Research Division, Mitsubishi Tanabe Pharma Corporation, Toda‑shi, Saitama 335‑8505, Japan
| | - Yasuhiro Egi
- Pharmacology Research Laboratories II, Research Division, Mitsubishi Tanabe Pharma Corporation, Toda‑shi, Saitama 335‑8505, Japan
| | - Hiroyuki Utsumi
- Safety Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Toda‑shi, Saitama 335‑8505, Japan
| | - Tetsuhiro Kakimoto
- Safety Research Laboratory, Research Division, Mitsubishi Tanabe Pharma Corporation, Toda‑shi, Saitama 335‑8505, Japan
| | - Kazuo Suzuki
- Pharmacology Research Laboratories II, Research Division, Mitsubishi Tanabe Pharma Corporation, Toda‑shi, Saitama 335‑8505, Japan
| |
Collapse
|
27
|
Han SJ, Kim JI, Park JW, Park KM. Hydrogen sulfide accelerates the recovery of kidney tubules after renal ischemia/reperfusion injury. Nephrol Dial Transplant 2015; 30:1497-1506. [DOI: 10.1093/ndt/gfv226] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
|
28
|
Jang HS, Padanilam BJ. Simultaneous deletion of Bax and Bak is required to prevent apoptosis and interstitial fibrosis in obstructive nephropathy. Am J Physiol Renal Physiol 2015; 309:F540-50. [PMID: 26180237 DOI: 10.1152/ajprenal.00170.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 07/13/2015] [Indexed: 12/14/2022] Open
Abstract
Proximal tubular injury and apoptosis are key mediators of the development of kidney fibrosis, a hallmark of chronic kidney disease. However, the molecular mechanism by which tubular apoptotic cell death leads to kidney fibrosis is poorly understood. In the present study, we tested the roles of Bcl-2-associated X (Bax) and Bcl-2 antagonist/killer (Bak), two crucial proteins involved in intrinsic apoptotic cell death, in the progression of kidney fibrosis. Mice with proximal tubule-specific Bax deletion, systemic deletion of Bak, and dual deletion of Bax and Bak were subjected to unilateral ureteral obstruction (UUO). Dual deficiency of Bax and Bak inhibited tubular apoptosis and atrophy. Consistent with decreased tubular injury, dual ablation of Bax and Bak suppressed UUO-induced inflammation and kidney fibrosis with decreased tubular cell cycle arrest, expression of fibrogenic and inflammatory cytokines, and oxidative stress in the kidney. Bax or Bak deficiency was insufficient to prevent apoptosis and all other aforementioned malevolent effects, suggesting compensatory mediation by each other in the respective signaling pathways. These data suggest that dual ablation of Bax and Bak in the kidney is required to prevent UUO-induced tubular apoptosis and the consequent kidney inflammation and fibrosis.
Collapse
Affiliation(s)
- Hee-Seong Jang
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and
| | - Babu J Padanilam
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; and Department of Internal Medicine, Section of Nephrology, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
29
|
Ding J, Ma Z, Liu H, Kwan P, Iwashina T, Shankowsky HA, Wong D, Tredget EE. The therapeutic potential of a C-X-C chemokine receptor type 4 (CXCR-4) antagonist on hypertrophic scarring in vivo. Wound Repair Regen 2015; 22:622-30. [PMID: 25139227 DOI: 10.1111/wrr.12208] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 07/12/2014] [Indexed: 12/17/2022]
Abstract
Effective prevention and treatment of hypertrophic scars (HTSs), a dermal form of fibrosis that frequently occurs following thermal injury to deep dermis, are unsolved significant clinical problems. Previously, we have found that stromal cell-derived factor 1/CXCR4 signaling is up-regulated during wound healing in burn patients and HTS tissue after thermal injury. We hypothesize that blood-borne mononuclear cells are recruited into wound sites after burn injury through the chemokine pathway of stromal cell-derived factor 1 and its receptor CXCR4. Deep dermal injuries to the skin are often accompanied by prolonged inflammation, which leads to chemotaxis of mononuclear cells into the wounds by chemokine signaling where fibroblast activation occurs and ultimately HTS are formed. Blocking mononuclear cell recruitment and fibroblast activation, CXCR4 antagonism is expected to reduce or minimize scar formation. In this study, the inhibitory effect of CXCR4 antagonist CTCE-9908 on dermal fibrosis was determined in vivo using a human HTS-like nude mouse model, in which split-thickness human skin is transplanted into full-thickness dorsal excisional wounds in athymic mice, where these wounds subsequently develop fibrotic scars that resemble human HTS as previously described. CTCE-9908 significantly attenuated scar formation and contraction, reduced the accumulation of macrophages and myofibroblasts, enhanced the remodeling of collagen fibers, and down-regulated the gene and protein expression of fibrotic growth factors in the human skin tissues. These findings support the potential therapeutic value of CXCR4 antagonist in dermal fibrosis and possibly other fibroproliferative disorders.
Collapse
Affiliation(s)
- Jie Ding
- Wound Healing Research Group, Division of Plastic and Reconstructive Surgery, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Origin of myofibroblasts and cellular events triggering fibrosis. Kidney Int 2014; 87:297-307. [PMID: 25162398 DOI: 10.1038/ki.2014.287] [Citation(s) in RCA: 271] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 04/04/2014] [Accepted: 04/10/2014] [Indexed: 01/13/2023]
Abstract
Renal fibrosis is a major hallmark of chronic kidney disease that is considered to be a common end point of various types of renal disease. To date, the biological meaning of fibrosis during the progression of chronic kidney diseases is unknown and possibly depends on the cell type contributing to extracellular matrix production. During the past decade, the origin of myofibroblasts in the kidney has been intensively investigated. Determining the origins of renal myofibroblasts is important because these might account for the heterogeneous characteristics and behaviors of myofibroblasts. Current data strongly suggest that collagen-producing myofibroblasts in the kidney can be derived from various cellular sources. Resident renal fibroblasts and cells of hematopoietic origin migrating into the kidney seem to be the most important ancestors of myofibroblasts. It is likely that both cell types communicate with each other and also with other cell types in the kidney. In this review, we will discuss the current knowledge on the origin of scar-producing myofibroblasts and cellular events triggering fibrosis.
Collapse
|
31
|
Jang HS, Kim JI, Noh M, Rhee MH, Park KM. Regulator of G protein signaling 2 (RGS2) deficiency accelerates the progression of kidney fibrosis. Biochim Biophys Acta Mol Basis Dis 2014; 1842:1733-41. [PMID: 24973550 DOI: 10.1016/j.bbadis.2014.06.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/02/2014] [Accepted: 06/18/2014] [Indexed: 01/07/2023]
Abstract
The regulator of G protein signaling 2 (RGS2) is a potent negative regulator of Gq protein signals including the angiotensin II (AngII)/AngII receptor signal, which plays a critical role in the progression of fibrosis. However, the role of RGS2 on the progression of kidney fibrosis has not been assessed. Here, we investigated the role of RGS2 in kidney fibrosis induced by unilateral ureteral obstruction (UUO) in mice. UUO resulted in increased expression of RGS2 mRNA and protein in the kidney along with increases of AngII and its type 1 receptor (AT1R) signaling and fibrosis. Furthermore, UUO increased the levels of F4/80, Ly6G, myeloperoxidase, and CXCR4 in the kidneys. RGS2 deficiency significantly enhanced these changes in the kidney. RGS2 deletion in the bone marrow-derived cells by transplanting the bone marrow of RGS2 knock-out mice into wild type mice enhanced UUO-induced kidney fibrosis. Overexpression of RGS2 in HEK293 cells, a human embryonic kidney cell line, and RAW264.7 cells, a monocyte/macrophage line, inhibited the AngII-induced activation of ERK and increase of CXCR4 expression. These findings provide the first evidence that RGS2 negatively regulates the progression of kidney fibrosis following UUO, likely by suppressing fibrogenic and inflammatory responses through the inhibition of AngII/AT1R signaling.
Collapse
Affiliation(s)
- Hee-Seong Jang
- Department of Anatomy, Cardiovascular Research Institute, BK21 Plus Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu 700-422, Republic of Korea
| | - Jee In Kim
- Department of Molecular Medicine and Obesity-Mediated Disease Research Center, College of Medicine, Keimyung University, Daegu, 704-701, Republic of Korea
| | - Mira Noh
- Department of Anatomy, Cardiovascular Research Institute, BK21 Plus Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu 700-422, Republic of Korea
| | - Man Hee Rhee
- Laboratory of Physiology and Signaling, College of Veterinary Medicine, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Kwon Moo Park
- Department of Anatomy, Cardiovascular Research Institute, BK21 Plus Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu 700-422, Republic of Korea.
| |
Collapse
|
32
|
Jang HS, Kim JI, Han SJ, Park KM. Recruitment and subsequent proliferation of bone marrow-derived cells in the postischemic kidney are important to the progression of fibrosis. Am J Physiol Renal Physiol 2014; 306:F1451-61. [PMID: 24740786 DOI: 10.1152/ajprenal.00017.2014] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Acute kidney injury (AKI) is an independent risk factor of the development of chronic kidney disease. Kidney fibrosis is a typical feature of chronic kidney disease and is characterized as an expansion of the interstitium due to increases in extracellular matrix molecules and interstitial cells caused by accumulations of extrarenal cells and by the proliferation or differentiation of intrarenal cells. However, the role of bone marrow-derived cells (BMDCs) in AKI-induced kidney fibrosis remains to be defined. Here, we investigated the role of BMDCs in kidney fibrosis after ischemia-reperfusion injury (IRI)-induced AKI in green fluorescent protein (GFP)-expressing bone marrow chimeric mice. IRI resulted in severe fibrotic changes in kidney tissues and dramatically increased interstitial cell numbers. Furthermore, GFP-expressing BMDCs accounted for >80% of interstitial cells in fibrotic kidneys. Interstitial GFP-expressing cells expressed α-smooth muscle actin (a myofibroblast marker), fibroblast-specific protein-1 (a fibroblast marker), collagen type III, and F4/80 (a macrophage marker). Over 20% of interstitial cells were bromodeoxyuridine-incorporating (proliferating) cells, and of these, 80% cells were GFP-expressing BMDCs. Daily treatment of IRI mice with apocynin (a NADPH oxidase inhibitor that functions as an antioxidant) from the day after surgery until euthanization slightly inhibited these changes with a small reduction of fibrosis. Taken together, our findings show that BMDCs make a major contribution to IRI-induced fibrosis due to their infiltration, subsequent differentiation, and proliferation in injured kidneys, suggesting that BMDCs be considered an important target for the treatment of kidney fibrosis.
Collapse
Affiliation(s)
- Hee-Seong Jang
- Department of Anatomy and BK21 Plus Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
| | - Jee In Kim
- Department of Molecular Medicine, Keimyung University School of Medicine, Daegu, Republic of Korea
| | - Sang Jun Han
- Department of Anatomy and BK21 Plus Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | - Kwon Moo Park
- Department of Anatomy and BK21 Plus Biomedical Convergence Program, Kyungpook National University School of Medicine, Daegu, Republic of Korea; Cardiovascular Research Institute, Kyungpook National University School of Medicine, Daegu, Republic of Korea; and
| |
Collapse
|
33
|
Yu YL, Yiang GT, Chou PL, Tseng HH, Wu TK, Hung YT, Lin PS, Lin SY, Liu HC, Chang WJ, Wei CW. Dual role of acetaminophen in promoting hepatoma cell apoptosis and kidney fibroblast proliferation. Mol Med Rep 2014; 9:2077-84. [PMID: 24682227 PMCID: PMC4055434 DOI: 10.3892/mmr.2014.2085] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 02/28/2014] [Indexed: 12/19/2022] Open
Abstract
Acetaminophen (APAP), is a safe analgesic and antipyretic drug at therapeutic dose, and is widely used in the clinic. However, high doses of APAP can induce hepatotoxicity and nephrotoxicity. Most studies have focused on high‑dose APAP‑induced acute liver and kidney injury. So far, few studies have investigated the effects of the therapeutic dose (1/10 of the high dose) or of the low dose (1/100 of the high dose) of APAP on the cells. The aim of this study was to investigate the cellular effects of therapeutic- or low‑dose APAP treatment on hepatoma cells and kidney fibroblasts. As expected, high‑dose APAP treatment inhibited while therapeutic and low‑dose treatment did not inhibit cell survival of kidney tubular epithelial cells. In addition, therapeutic-dose treatment induced an increase in the H2O2 level, activated the caspase‑9/‑3 cascade, and induced cell apoptosis of hepatoma cells. Notably, APAP promoted fibroblast proliferation, even at low doses. This study demonstrates that different cellular effects are exerted upon treatment with different APAP concentrations. Our results indicate that treatment with the therapeutic dose of APAP may exert an antitumor activity on hepatoma, while low‑dose treatment may be harmful for patients with fibrosis, since it may cause proliferation of fibroblasts.
Collapse
Affiliation(s)
- Yung-Luen Yu
- Graduate Institute of Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan, R.O.C
| | - Pei-Lun Chou
- Division of Allergy‑Immunology‑Rheumatology, Department of Internal Medicine, Saint Mary's Hospital Luodong, Yilan 265, Taiwan, R.O.C
| | - Hsu-Hung Tseng
- Division of General Surgery, Taichung Hospital, Ministry of Health and Welfare, Taichung 403, Taiwan, R.O.C
| | - Tsai-Kun Wu
- 2The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Yu-Ting Hung
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Taichung 433, Taiwan, R.O.C
| | - Pei-Shiuan Lin
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Taichung 433, Taiwan, R.O.C
| | - Shu-Yu Lin
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Taichung 433, Taiwan, R.O.C
| | - Hsiao-Chun Liu
- Department of Nursing, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei 231, Taiwan, R.O.C
| | - Wei-Jung Chang
- Graduate Institute of Cancer Biology and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan, R.O.C
| | - Chyou-Wei Wei
- Department of Nutrition, Master Program of Biomedical Nutrition, Hungkuang University, Taichung 433, Taiwan, R.O.C
| |
Collapse
|
34
|
Activation of ERK accelerates repair of renal tubular epithelial cells, whereas it inhibits progression of fibrosis following ischemia/reperfusion injury. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1998-2008. [DOI: 10.1016/j.bbadis.2013.07.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 06/12/2013] [Accepted: 07/01/2013] [Indexed: 11/18/2022]
|
35
|
Jung KJ, Jang HS, Kim JI, Han SJ, Park JW, Park KM. Involvement of hydrogen sulfide and homocysteine transsulfuration pathway in the progression of kidney fibrosis after ureteral obstruction. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1989-97. [PMID: 23846016 DOI: 10.1016/j.bbadis.2013.06.015] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 05/22/2013] [Accepted: 06/26/2013] [Indexed: 12/16/2022]
Abstract
Hydrogen sulfide (H2S) produced by cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE) in the transsulfuration pathway of homocysteine plays a number of pathophysiological roles. Hyperhomocysteinemia is involved in kidney fibrosis. However, the role of H2S in kidney fibrosis remains to be defined. Here, we investigated the role of H2S and its acting mechanism in unilateral ureteral obstruction (UO)-induced kidney fibrosis in mice. UO decreased expressions of CBS and CSE in the kidney with decrease of H2S concentration. Treatment with sodium hydrogen sulfide (NaHS, a H2S producer) during UO reduced UO-induced oxidative stress with preservations of catalase, copper-zinc superoxide dismutase (CuZnSOD), and manganese superoxide dismutase (MnSOD) expression, and glutathione level. In addition, NaHS mitigated decreases of CBS and CSE expressions, and H2S concentration in the kidney. NaHS treatment attenuated UO-induced increases in levels of TGF-β1, activated Smad3, and activated NF-κB. This study provided the first evidence of involvement of the transsulfuration pathway and H2S in UO-induced kidney fibrosis, suggesting that H2S and its transsulfuration pathway may be a potential target for development of therapeutics for fibrosis-related diseases.
Collapse
Affiliation(s)
- Kyong-Jin Jung
- Department of Anatomy, Kyungpook National University School of Medicine, Daegu 700-422, Republic of Korea
| | | | | | | | | | | |
Collapse
|
36
|
Kim JI, Jang HS, Jeong JH, Noh MR, Choi JY, Park KM. Defect in Runx2 gene accelerates ureteral obstruction-induced kidney fibrosis via increased TGF-β signaling pathway. Biochim Biophys Acta Mol Basis Dis 2013; 1832:1520-7. [PMID: 23639629 DOI: 10.1016/j.bbadis.2013.04.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2012] [Revised: 04/17/2013] [Accepted: 04/22/2013] [Indexed: 02/07/2023]
Abstract
Runt-related transcription factor 2 (Runx2) plays an important role in bone formation and de novo synthesis of proteins, including type 1 collagen. Runx2 has a potent effect on signaling of transforming growth factor (TGF)-β and vice versa, implicating its significant role in fibrosis. Chronic renal failure comprises fibrosis, characterized as an increase in TGF-β signaling, and expression of α-smooth muscle actin (α-SMA), and extracellular matrix proteins. Here, we evaluated the role of Runx2 in ureteral obstruction (UO)-induced kidney fibrosis using mice whose Runx2 gene expression is genetically down-regulated. UO caused tubular atrophy and dilation, expansion of interstitium, and increased expression of collagens and α-SMA with a concomitant decrease in expression of Runx2. Deficiency of Runx2 gene (Runx2(+/-) mice) showed higher expression of collagens and α-SMA in the kidney following UO compared to wild type (Runx2(+/+)) mice. UO-induced activation of TGF-β signaling was higher in the Runx2(+/-) kidney than Runx2(+/+) kidney, suggesting an inhibitory effect of Runx2 on TGF-β signaling in kidney fibrosis. Besides, overexpression of the Runx2 gene using an adenoviral vector in kidney tubule cells resulted in attenuated TGF-β-induced Smad3 phosphorylation and expressions of α-SMA and collagen I. Furthermore, Runx2 gene deficient mouse embryonic fibroblasts induced greater activation of Smad3 and expression of α-SMA in response to TGF-β. Collectively, Runx2 plays a protective role in UO-induced kidney fibrosis by inhibition of TGF-β signaling, suggesting Runx2 as a novel target for protection against fibrosis-related diseases such as chronic renal failure.
Collapse
Affiliation(s)
- Jee In Kim
- Department of Anatomy and BK21, Kyungpook National University School of Medicine, Republic of Korea
| | | | | | | | | | | |
Collapse
|