1
|
Sijbesma JWA, van Waarde A, Klooster A, Kion I, Slart RHJA, Lammertsma AA, Giacobbo BL, Boersma HH, Dierckx RAJO, van Goor H, Bakker SJL. Caloric restriction reduces proteinuria in male rats with established nephropathy. Physiol Rep 2024; 12:e15942. [PMID: 38439743 PMCID: PMC10912948 DOI: 10.14814/phy2.15942] [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: 12/06/2023] [Revised: 01/15/2024] [Accepted: 01/15/2024] [Indexed: 03/06/2024] Open
Abstract
Reducing proteinuria is a crucial approach in preventing kidney function loss. Previous preclinical studies indicated that caloric restriction (CR) imposed at a young age protects against age-related proteinuria. However, these studies have not explored CR in established renal disease. Therefore, this study aimed to investigate the impact of CR on established proteinuria. Rats, aged 12 ± 2 weeks, were administered 2.1 mg/kg of Adriamycin. Six weeks after injection, protein excretion was measured, and a [13 N]ammonia positron emission tomography (PET) scan was conducted to assess kidney perfusion. After 7 weeks rats were divided into four groups: ad libitum (AL) and CR groups fed either a 12% or a 20% protein diet. All groups were treated for 12 weeks. Blood pressure was measured and a second PET scan was acquired at the end of the study. The animals subjected to CR exhibited a 20.3% decrease in protein excretion (p = 0.003) compared to those in the AL groups. Additionally, blood pressure in the CR group was 21.2% lower (p < 0.001) than in the AL groups. While kidney function declined over time in all groups, the 20% CR group demonstrated the smallest decline. Thus CR effectively reduces urinary protein excretion and lowers blood pressure in rats with established proteinuria.
Collapse
Affiliation(s)
- J. W. A. Sijbesma
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - A. van Waarde
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - A. Klooster
- Department of PathologyPathologie FrieslandLeeuwardenThe Netherlands
| | - I. Kion
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - R. H. J. A. Slart
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
- Department of Biomedical Photonic Imaging, Faculty of Science and TechnologyUniversity of TwenteEnschedeThe Netherlands
| | - A. A. Lammertsma
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - B. Lima Giacobbo
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - H. H. Boersma
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
- Department of Clinical Pharmacy and PharmacologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - R. A. J. O. Dierckx
- Department of Nuclear Medicine and Molecular ImagingUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - H. van Goor
- Department of Pathology and Medical BiologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| | - S. J. L. Bakker
- Department of NephrologyUniversity Medical Center Groningen, University of GroningenGroningenThe Netherlands
| |
Collapse
|
2
|
Peng Z, Wang H, Zheng J, Wang J, Xiang Y, Liu C, Ji M, Liu H, Pan L, Qin X, Qu X. Is the proximal tubule the focus of tubulointerstitial fibrosis? Heliyon 2023; 9:e13508. [PMID: 36846656 PMCID: PMC9950842 DOI: 10.1016/j.heliyon.2023.e13508] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/15/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Tubulointerstitial fibrosis (TIF), a common end result of almost all progressive chronic kidney diseases (CKD), is also the best predictor of kidney survival. Almost all cells in the kidney are involved in the progression of TIF. Myofibroblasts, the primary producers of extracellular matrix, have previously received a great deal of attention; however, a large body of emerging evidence reveals that proximal tubule (PT) plays a central role in TIF progression. In response to injury, renal tubular epithelial cells (TECs) transform into inflammatory and fibroblastic cells, producing various bioactive molecules that drive interstitial inflammation and fibrosis. Here we reviewed the increasing evidence for the key role of the PT in promoting TIF in tubulointerstitial and glomerular injury and discussed the therapeutic targets and carrier systems involving the PT that holds particular promise for treating patients with fibrotic nephropathy.
Collapse
Affiliation(s)
- Zhi Peng
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Hui Wang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Jiaoyun Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jie Wang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Yang Xiang
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Chi Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Ming Ji
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Huijun Liu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Lang Pan
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Xiaoqun Qin
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| | - Xiangping Qu
- Department of Physiology, School of Basic Medical Science, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
3
|
Steichen C, Hervé C, Hauet T, Bourmeyster N. Rho GTPases in kidney physiology and diseases. Small GTPases 2022; 13:141-161. [PMID: 34138686 PMCID: PMC9707548 DOI: 10.1080/21541248.2021.1932402] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 02/06/2023] Open
Abstract
Rho family GTPases are molecular switches best known for their pivotal role in dynamic regulation of the actin cytoskeleton, but also of cellular morphology, motility, adhesion and proliferation. The prototypic members of this family (RhoA, Rac1 and Cdc42) also contribute to the normal kidney function and play important roles in the structure and function of various kidney cells including tubular epithelial cells, mesangial cells and podocytes. The kidney's vital filtration function depends on the structural integrity of the glomerulus, the proximal portion of the nephron. Within the glomerulus, the architecturally actin-based cytoskeleton podocyte forms the final cellular barrier to filtration. The glomerulus appears as a highly dynamic signalling hub that is capable of integrating intracellular cues from its individual structural components. Dynamic regulation of the podocyte cytoskeleton is required for efficient barrier function of the kidney. As master regulators of actin cytoskeletal dynamics, Rho GTPases are therefore of critical importance for sustained kidney barrier function. Dysregulated activities of the Rho GTPases and of their effectors are implicated in the pathogenesis of both hereditary and idiopathic forms of kidney diseases. Diabetic nephropathy is a progressive kidney disease that is caused by injury to kidney glomeruli. High glucose activates RhoA/Rho-kinase in mesangial cells, leading to excessive extracellular matrix production (glomerulosclerosis). This RhoA/Rho-kinase pathway also seems involved in the post-transplant hypertension frequently observed during treatment with calcineurin inhibitors, whereas Rac1 activation was observed in post-transplant ischaemic acute kidney injury.
Collapse
Affiliation(s)
- Clara Steichen
- Inserm UMR-1082 Irtomit, Poitiers, France
- Faculté De Médecine Et De Pharmacie, Université De Poitiers, Poitiers, France
| | | | - Thierry Hauet
- Inserm UMR-1082 Irtomit, Poitiers, France
- Faculté De Médecine Et De Pharmacie, Université De Poitiers, Poitiers, France
- Department of Medical Biology, Service De Biochimie, CHU De Poitiers, Poitiers, France
| | - Nicolas Bourmeyster
- Faculté De Médecine Et De Pharmacie, Université De Poitiers, Poitiers, France
- Department of Medical Biology, Service De Biochimie, CHU De Poitiers, Poitiers, France
- Laboratoire STIM CNRS ERL 7003, Université de Poitiers, Poitiers Cédex, France
| |
Collapse
|
4
|
Zheng XP, Nie Q, Feng J, Fan XY, Jin YL, Chen G, Du JW. Kidney-targeted baicalin-lysozyme conjugate ameliorates renal fibrosis in rats with diabetic nephropathy induced by streptozotocin. BMC Nephrol 2020; 21:174. [PMID: 32398108 PMCID: PMC7216346 DOI: 10.1186/s12882-020-01833-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/29/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is one of the most common and serious complications of diabetes, and is the most important cause of death for diabetic patients. Baicalin (BAI) has anti-oxidative, anti-inflammatory and anti-apoptotic activities, which play a role in attenuating insulin resistance and protecting the kidney. Moreover, cell-specific targeting of renal tubular cells is an approach to enhance drug accumulation in the kidney. METHODS Forty-five Sprague-Dawley rats were divided into four groups. A diabetes model was created using streptozotocin (STZ) intraperitoneally injection. The four groups included: Control group (n = 10), DN (n = 15), BAI treatment (BAI; n = 10) and BAI-LZM treatment (BAI-LZM; n = 10) groups. In the current study, the renoprotection and anti-fibrotic effects of BAI-lysozyme (LZM) conjugate were further investigated in rats with DN induced by STZ compared with BAI treatment alone. RESULTS The results suggest that BAI-LZM better ameliorates renal impairment, metabolic disorder and renal fibrosis than BAI alone in rats with DN, and the potential regulatory mechanism likely involves inhibiting inflammation via the nuclear factor-κB signaling pathway, inhibiting extracellular matrix accumulation via the transforming growth factor-β/Smad3 pathway and regulating cell proliferation via the insulin-like growth factor (IGF)-1/IGF-1 receptor/p38 Mitogen-activated protein kinase (MAPK) pathway. BAI and the kidney-targeted BAI-LZM can utilize the body's cytoprotective pathways to reactivate autophagy (as indicated by the autophagy markers mechanistic target of rapamycin and sirtuin 1 to ameliorate DN outcomes. CONCLUSIONS Our data support the traditional use of S. baicalensis as an important anti-DN traditional chinese medicine (TCM), and BAI, above all BAI-LZM, is a promising source for the identification of molecules with anti-DN effects.
Collapse
Affiliation(s)
- Xiao-Peng Zheng
- Department of basic medical sciences, Taizhou University hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, 318000, China
- College of Basic Medical Sciences, Jiamusi University, No 148 Xuefu Street, Jiamusi, 154007, China
| | - Qing Nie
- Weifang centers for disease control and prevention, No 4801 Huixian Road, Gaoxin Distric, Weifang, 261061, Shandong Province, China
| | - Jing Feng
- Department of basic medical sciences, Taizhou University hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, 318000, China
- College of Basic Medical Sciences, Jiamusi University, No 148 Xuefu Street, Jiamusi, 154007, China
| | - Xiao-Yan Fan
- Department of basic medical sciences, Taizhou University hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, 318000, China
| | - Yue-Lei Jin
- Department of basic medical sciences, Taizhou University hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, 318000, China
| | - Guang Chen
- Department of basic medical sciences, Taizhou University hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, 318000, China.
| | - Ji-Wei Du
- Nursing department, Xiang'An Hospital, Xiamen University, Xiamen, 361005, China.
| |
Collapse
|
5
|
Jafree DJ, Long DA. Beyond a Passive Conduit: Implications of Lymphatic Biology for Kidney Diseases. J Am Soc Nephrol 2020; 31:1178-1190. [PMID: 32295825 DOI: 10.1681/asn.2019121320] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The kidney contains a network of lymphatic vessels that clear fluid, small molecules, and cells from the renal interstitium. Through modulating immune responses and via crosstalk with surrounding renal cells, lymphatic vessels have been implicated in the progression and maintenance of kidney disease. In this Review, we provide an overview of the development, structure, and function of lymphatic vessels in the healthy adult kidney. We then highlight the contributions of lymphatic vessels to multiple forms of renal pathology, emphasizing CKD, transplant rejection, and polycystic kidney disease and discuss strategies to target renal lymphatics using genetic and pharmacologic approaches. Overall, we argue the case for lymphatics playing a fundamental role in renal physiology and pathology and treatments modulating these vessels having therapeutic potential across the spectrum of kidney disease.
Collapse
Affiliation(s)
- Daniyal J Jafree
- Developmental Biology and Cancer Programme, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom.,MB/PhD Programme, Faculty of Medical Sciences, University College London, London, United Kingdom
| | - David A Long
- Developmental Biology and Cancer Programme, Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
6
|
Campeiro JD, Dam W, Monte GG, Porta LC, Oliveira LCGD, Nering MB, Viana GM, Carapeto FC, Oliveira EB, van den Born J, Hayashi MAF. Long term safety of targeted internalization of cell penetrating peptide crotamine into renal proximal tubular epithelial cells in vivo. Sci Rep 2019; 9:3312. [PMID: 30824773 PMCID: PMC6397221 DOI: 10.1038/s41598-019-39842-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 01/21/2019] [Indexed: 02/07/2023] Open
Abstract
Activated proximal tubular epithelial cells (PTECs) play a crucial role in progressive tubulo-interstitial fibrosis in native and transplanted kidneys. Targeting PTECs by non-viral delivery vectors might be useful to influence the expression of important genes and/or proteins in order to slow down renal function loss. However, no clinical therapies that specifically target PTECs are available at present. We earlier showed that a cationic cell penetrating peptide isolated from South American rattlesnake venom, named crotamine, recognizes cell surface heparan sulfate proteoglycans and accumulates in cells. In healthy mice, crotamine accumulates mainly in kidneys after intraperitoneal (ip) injection. Herein we demonstrate for the first time, the overall safety of acute or long-term treatment with daily ip administrated crotamine for kidneys functions. Accumulation of ip injected crotamine in the kidney brush border zone of PTECs, and its presence inside these cells were observed. In addition, significant lower in vitro crotamine binding, uptake and reporter gene transport and expression could be observed in syndecan-1 deficient HK-2 PTECs compared to wild-type cells, indicating that the absence of syndecan-1 impairs crotamine uptake into PTECs. Taken together, our present data show the safety of in vivo long-term treatment with crotamine, and its preferential uptake into PTECs, which are especially rich in HSPGs such as syndecan-1. In addition to the demonstrated in vitro gene delivery mediated by crotamine in HK-2 cells, the potential applicability of crotamine as prototypic non-viral (gene) delivery nanocarrier to modulate PTEC gene and/or protein expression was confirmed.
Collapse
Affiliation(s)
- Joana Darc Campeiro
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Wendy Dam
- Department Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Gabriela Guilherme Monte
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Lucas Carvalho Porta
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | | | - Marcela Bego Nering
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Gustavo Monteiro Viana
- Departamento de Bioquímica, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Fernando Cintra Carapeto
- Departamento de Patologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil
| | - Eduardo Brandt Oliveira
- Departamento de Bioquímica e Imunologia, Universidade de São Paulo (USP-FMRP), Ribeirão Preto, Brazil
| | - Jacob van den Born
- Department Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Mirian A F Hayashi
- Departamento de Farmacologia, Escola Paulista de Medicina (EPM), Universidade Federal de São Paulo (UNIFESP), São Paulo, SP, Brazil.
| |
Collapse
|
7
|
Yamamoto T, Ugawa Y, Kawamura M, Yamashiro K, Kochi S, Ideguchi H, Takashiba S. Modulation of microenvironment for controlling the fate of periodontal ligament cells: the role of Rho/ROCK signaling and cytoskeletal dynamics. J Cell Commun Signal 2018; 12:369-378. [PMID: 29086204 PMCID: PMC5842188 DOI: 10.1007/s12079-017-0425-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 10/17/2017] [Indexed: 12/20/2022] Open
Abstract
Cells behave in a variety of ways when they perceive changes in their microenvironment; the behavior of cells is guided by their coordinated interactions with growth factors, niche cells, and extracellular matrix (ECM). Modulation of the microenvironment affects the cell morphology and multiple gene expressions. Rho/Rho-associated coiled-coil-containing protein kinase (ROCK) signaling is one of the key regulators of cytoskeletal dynamics and actively and/or passively determines the cell fate, such as proliferation, migration, differentiation, and apoptosis, by reciprocal communication with the microenvironment. During periodontal wound healing, it is important to recruit the residential stem cells into the defect site for regeneration and homeostasis of the periodontal tissue. Periodontal ligament (PDL) cells contain a heterogeneous fibroblast population, including mesenchymal stem cells, and contribute to the reconstruction of tooth-supporting tissues. Therefore, bio-regeneration of PDL cells has been the ultimate goal of periodontal therapy for decades. Recent stem cell researches have shed light on intrinsic ECM properties, providing paradigm shifts in cell fate determination. This review focuses on the role of ROCK activity and the effects of Y-27632, a specific inhibitor of ROCK, in the modulation of ECM-microenvironment. Further, it presents the current understanding of how Rho/ROCK signaling affects the fate determination of stem cells, especially PDL cells. In addition, we have also discussed in detail the underlying mechanisms behind the reciprocal response to the microenvironment.
Collapse
Affiliation(s)
- Tadashi Yamamoto
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Yuki Ugawa
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Mari Kawamura
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Keisuke Yamashiro
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Shinsuke Kochi
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Hidetaka Ideguchi
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan
| | - Shogo Takashiba
- Department of Pathophysiology - Periodontal Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8525, Japan.
| |
Collapse
|
8
|
Kimura Y, Sumiyoshi M. Resveratrol Prevents Tumor Growth and Metastasis by Inhibiting Lymphangiogenesis and M2 Macrophage Activation and Differentiation in Tumor-associated Macrophages. Nutr Cancer 2016; 68:667-78. [PMID: 27145432 DOI: 10.1080/01635581.2016.1158295] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Antitumor and antimetastatic effects of resveratrol on tumor-induced lymphangiogenesis through the regulation of M2 macrophages in tumor-associated macrophages currently remain unknown. Therefore, we herein examined the effects of resveratrol on M2 macrophage activation and differentiation, and those of resveratrol-treated condition medium (CM) in M2 macrophages on vascular endothelial cell growth factor (VEGF)-C-induced migration, invasion, and tube formation by human lymphatic endothelial cells (HLECs). Resveratrol (50 μM or 5-50 μM) inhibited the production of interleukin-10 and monocyte chemoattractant protein-1 in M2 macrophages, whereas it promoted that of transforming growth factor-β1. Resveratrol (25 and 50 μM) inhibited the phosphorylation of signal transducer and activator of transcript 3 without affecting its expression in the differentiation process of M2 macrophages. Furthermore, resveratrol-treated CM of M2 macrophages inhibited VEGF-C-induced HLEC migration, invasion, and lymphangiogenesis. Resveratrol (25 mg/kg, twice daily) inhibited tumor growth and metastasis to the lung and also reduced the area of lymphatic endothelial cells in tumors (in vivo). These results suggest that the antitumor and antimetastatic effects of resveratrol were partly due to antilymphangiogenesis through the regulation of M2 macrophage activation and differentiation.
Collapse
Affiliation(s)
- Yoshiyuki Kimura
- a Division of Biochemical Pharmacology, Department of Basic Medical Research, Ehime University Graduate School of Medicine , Toon City , Ehime , Japan
| | - Maho Sumiyoshi
- a Division of Biochemical Pharmacology, Department of Basic Medical Research, Ehime University Graduate School of Medicine , Toon City , Ehime , Japan
| |
Collapse
|
9
|
Lee W, Miyagawa Y, Long C, Zhang M, Cooper DKC, Hara H. Effect of Rho-kinase Inhibitor, Y27632, on Porcine Corneal Endothelial Cell Culture, Inflammation and Immune Regulation. Ocul Immunol Inflamm 2015; 24:579-93. [PMID: 26471144 DOI: 10.3109/09273948.2015.1056534] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE To investigate the effect of the Rho-kinase inhibitor, Y27632, on pig corneal endothelial cell (pCEC) culture, and on inflammation and immune regulation of the responses of human cells to pCECs. METHODS pCECs were cultured with/without Y27632 to assess cell proliferation and in vitro wound healing assay. The level of MCP-1 and VEGF in pCECs stimulated with human TNF-α were measured. Proliferation of human PBMCs stimulated with pCECs, and cytokine production in human T cells, and monocyte migration after stimulation were investigated. RESULTS Y27632 promoted pCEC proliferation, prevented pCEC death, and enhanced in vitro wound healing. After stimulation, there were significantly lower levels of MCP-1 and VEGF measured in pCECs cultured with Y27632, and significantly reduced human PBMC proliferation, cytokine production, and monocyte migration. CONCLUSIONS The application of the Rho-kinase inhibitor will be beneficial when culturing pCECs, and may provide a novel therapy to reduce inflammation after corneal xenotransplantation.
Collapse
Affiliation(s)
- Whayoung Lee
- a Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh , Pittsburgh , Pennsylvania , USA
| | - Yuko Miyagawa
- a Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh , Pittsburgh , Pennsylvania , USA
| | - Cassandra Long
- a Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh , Pittsburgh , Pennsylvania , USA
| | - Matthew Zhang
- a Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh , Pittsburgh , Pennsylvania , USA
| | - David K C Cooper
- a Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh , Pittsburgh , Pennsylvania , USA
| | - Hidetaka Hara
- a Thomas E. Starzl Transplantation Institute, Department of Surgery, University of Pittsburgh , Pittsburgh , Pennsylvania , USA
| |
Collapse
|
10
|
Yazdani S, Hijmans RS, Poosti F, Dam W, Navis G, van Goor H, van den Born J. Targeting tubulointerstitial remodeling in proteinuric nephropathy in rats. Dis Model Mech 2015; 8:919-30. [PMID: 26035383 PMCID: PMC4527281 DOI: 10.1242/dmm.018580] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Accepted: 05/07/2015] [Indexed: 12/23/2022] Open
Abstract
Proteinuria is an important cause of tubulointerstitial damage. Anti-proteinuric interventions are not always successful, and residual proteinuria often leads to renal failure. This indicates the need for additional treatment modalities by targeting the harmful downstream consequences of proteinuria. We previously showed that proteinuria triggers renal lymphangiogenesis before the onset of interstitial inflammation and fibrosis. However, the interrelationship of these interstitial events in proteinuria is not yet clear. To this end, we specifically blocked lymphangiogenesis (anti-VEGFR3 antibody), monocyte/macrophage influx (clodronate liposomes) or lymphocyte and myofibroblast influx (S1P agonist FTY720) separately in a rat model to investigate the role and the possible interaction of each of these phenomena in tubulointerstitial remodeling in proteinuric nephropathy. Proteinuria was induced in 3-month old male Wistar rats by adriamycin injection. After 6 weeks, when proteinuria has developed, rats were treated for another 6 weeks by anti-VEGFR3 antibody, clodronate liposomes or FTY720 up to week 12. In proteinuric rats, lymphangiogenesis, influx of macrophages, T cells and myofibroblasts, and collagen III deposition and interstitial fibrosis significantly increased at week 12 vs week 6. Anti-VEGFR3 antibody prevented lymphangiogenesis in proteinuric rats, however, without significant effects on inflammatory and fibrotic markers or proteinuria. Clodronate liposomes inhibited macrophage influx and partly reduced myofibroblast expression; however, neither significantly prevented the development of lymphangiogenesis, nor fibrotic markers and proteinuria. FTY720 prevented myofibroblast accumulation, T-cell influx and interstitial fibrosis, and partially reduced macrophage number and proteinuria; however, it did not significantly influence lymphangiogenesis and collagen III deposition. This study showed that proteinuria-induced interstitial fibrosis cannot be halted by blocking lymphangiogenesis or the influx of macrophages. On the other hand, FTY720 treatment did prevent T-cell influx, myofibroblast accumulation and interstitial fibrosis, but not renal lymphangiogenesis and proteinuria. We conclude that tubulointerstitial fibrosis and inflammation are separate from lymphangiogenesis, at least under proteinuric conditions. Summary: Targeting lymphangiogenesis, inflammation or fibrosis separately in a rat model of proteinuric nephropathy showed that treating any of these changes alone is not effective in treating the disease.
Collapse
Affiliation(s)
- Saleh Yazdani
- Department of Medicine, Division of Nephrology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Ryanne S Hijmans
- Department of Medicine, Division of Nephrology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Fariba Poosti
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Wendy Dam
- Department of Medicine, Division of Nephrology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Gerjan Navis
- Department of Medicine, Division of Nephrology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Harry van Goor
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Jacob van den Born
- Department of Medicine, Division of Nephrology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
11
|
Ramos AM, González-Guerrero C, Sanz A, Sanchez-Niño MD, Rodríguez-Osorio L, Martín-Cleary C, Fernández-Fernández B, Ruiz-Ortega M, Ortiz A. Designing drugs that combat kidney damage. Expert Opin Drug Discov 2015; 10:541-56. [PMID: 25840605 DOI: 10.1517/17460441.2015.1033394] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Kidney disease remains one of the last worldwide frontiers in the field of non-communicable human disease. From 1990 to 2013, chronic kidney disease (CKD) was the top non-communicable cause of death with a greatest increase in global years of life lost while mortality of acute kidney injury (AKI) still hovers around 50%. This reflects the paucity (for CKD) or lack of (for AKI) therapeutic approaches beyond replacing renal function. Understanding what the barriers are and what potential pathways may facilitate the design of new drugs to combat kidney disease is a key public health priority. AREAS COVERED The authors discuss the hurdles and opportunities for future drug development for kidney disease in light of experience accumulated with drugs that made it to clinical trials. EXPERT OPINION Inflammation, cell death and fibrosis are key therapeutic targets to combat kidney damage. While the specific targeting of drugs to kidney cells would be desirable, the technology is only working at the preclinical stage and with mixed success. Nanomedicines hold promise in this respect. Most drugs undergoing clinical trials for kidney disease have been repurposed from other indications. Currently, the chemokine receptor inhibitor CCX140 holds promise for CKD and the p53 inhibitor QPI-1002 for AKI.
Collapse
Affiliation(s)
- Adrián M Ramos
- Instituto de Investigación Sanitaria-Fundación Jiménez Díaz (IIS-FJD), Laboratory of Renal and Vascular Pathology and Diabetes , Av. Reyes Católicos 2, 28040, Madrid , Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Poosti F, Bansal R, Yazdani S, Prakash J, Post E, Klok P, van den Born J, de Borst MH, van Goor H, Poelstra K, Hillebrands JL. Selective delivery of IFN-γ to renal interstitial myofibroblasts: a novel strategy for the treatment of renal fibrosis. FASEB J 2015; 29:1029-42. [PMID: 25466892 DOI: 10.1096/fj.14-258459] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2023]
Abstract
Renal fibrosis leads to end-stage renal disease demanding renal replacement therapy because no adequate treatment exists. IFN-γ is an antifibrotic cytokine that may attenuate renal fibrosis. Systemically administered IFN-γ causes side effects that may be prevented by specific drug targeting. Interstitial myofibroblasts are the effector cells in renal fibrogenesis. Here, we tested the hypothesis that cell-specific delivery of IFN-γ to platelet-derived growth factor receptor β (PDGFRβ)-expressing myofibroblasts attenuates fibrosis in an obstructive nephropathy [unilateral ureteral obstruction (UUO)] mouse model. PEGylated IFN-γ conjugated to PDGFRβ-recognizing peptide [(PPB)-polyethylene glycol (PEG)-IFN-γ] was tested in vitro and in vivo for antifibrotic properties and compared with free IFN-γ. PDGFRβ expression was >3-fold increased (P < 0.05) in mouse fibrotic UUO kidneys and colocalized with α-smooth muscle actin-positive (SMA(+)) myofibroblasts. In vitro, PPB-PEG-IFN-γ significantly inhibited col1a1, col1a2, and α-SMA mRNA expression in TGF-β-activated NIH3T3 fibroblasts (P < 0.05). In vivo, PPB-PEG-IFN-γ specifically accumulated in PDGFRβ-positive myofibroblasts. PPB-PEG-IFN-γ treatment significantly reduced renal collagen I, fibronectin, and α-SMA mRNA and protein expression. Compared with vehicle treatment, PPB-PEG-IFN-γ preserved tubular morphology, reduced interstitial T-cell infiltration, and attenuated lymphangiogenesis (all P < 0.05) without affecting peritubular capillary density. PPB-PEG-IFN-γ reduced IFN-γ-related side effects as manifested by reduced major histocompatibility complex class II expression in brain tissue (P < 0.05 vs. free IFN-γ). Our findings demonstrate that specific targeting of IFN-γ to PDGFRβ-expressing myofibroblasts attenuates renal fibrosis and reduces systemic adverse effects.
Collapse
Affiliation(s)
- Fariba Poosti
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Saleh Yazdani
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Jai Prakash
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Eduard Post
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Pieter Klok
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Jacob van den Born
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Martin H de Borst
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Harry van Goor
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Klaas Poelstra
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| | - Jan-Luuk Hillebrands
- *Department of Pathology and Medical Biology, Division of Pathology, Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, and Department of Pharmacokinetics, Toxicology and Targeting, University of Groningen, Groningen, The Netherlands; and MIRA Institute, University of Twente, Enschede, The Netherlands
| |
Collapse
|
13
|
Hu H, Chen W, Ding J, Jia M, Yin J, Guo Z. Fasudil prevents calcium oxalate crystal deposit and renal fibrogenesis in glyoxylate-induced nephrolithic mice. Exp Mol Pathol 2015; 98:277-85. [PMID: 25697583 DOI: 10.1016/j.yexmp.2015.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 02/12/2015] [Accepted: 02/12/2015] [Indexed: 01/30/2023]
Abstract
Nephrolithiasis is a common kidney disease and one of the major causes of chronic renal insufficiency. We develop and utilize a glyoxylate induced mouse model of kidney calcium oxalate crystal deposition for studying the pharmacological effects of fasudil, a Rho associated protein kinase (ROCK) specific inhibitor, on the kidney injury and fibrosis caused by calcium oxalate crystallization and deposition. Glyoxylate was administrated intraperitoneally to C57BL/6J mice for five consecutive days to establish a mouse model of kidney calcium oxalate crystal formation and deposition. The results showed that the protein expression levels of E-cad and Pan-ck were lower, and the protein expression levels of α-SMA and Vim were higher, in the kidney tissue of the glyoxylate induced model mice compared with the control mice. The changes in protein expression were weakened when the animals were pretreated with fasudil before glyoxylate administration. Expression of ROCK, PAI-1, and p-Smad proteins in the kidney tissue increased in response to glyoxylate treatment, and the increase was eased when the animals were pretreated with fasudil. Expression of Smad2 and Smad3 in the kidney tissue remained unchanged after glyoxylate administration. Cell apoptosis and proliferation in the kidney cortex and medulla were enhanced in response to the glyoxylate induced calcium oxalate crystal formation and deposition, and fasudil pre-treatment was able to attenuate the enhancement. The results suggest that Fasudil reduces the glyoxylate induced kidney calcium crystal formation and deposition and slows down the kidney fibrogenesis caused by calcium crystal deposition. The possible mechanism may be related the regulatory effects on Rho/ROCK signal transduction and epithelial-mesenchymal transition (EMT).
Collapse
Affiliation(s)
- Haiyan Hu
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Wei Chen
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Jiarong Ding
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Meng Jia
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Jingjing Yin
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China
| | - Zhiyong Guo
- Department of Nephrology, Changhai Hospital, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
14
|
Knipe RS, Tager AM, Liao JK. The Rho kinases: critical mediators of multiple profibrotic processes and rational targets for new therapies for pulmonary fibrosis. Pharmacol Rev 2015; 67:103-17. [PMID: 25395505 PMCID: PMC4279074 DOI: 10.1124/pr.114.009381] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is characterized by progressive lung scarring, short median survival, and limited therapeutic options, creating great need for new pharmacologic therapies. IPF is thought to result from repetitive environmental injury to the lung epithelium, in the context of aberrant host wound healing responses. Tissue responses to injury fundamentally involve reorganization of the actin cytoskeleton of participating cells, including epithelial cells, fibroblasts, endothelial cells, and macrophages. Actin filament assembly and actomyosin contraction are directed by the Rho-associated coiled-coil forming protein kinase (ROCK) family of serine/threonine kinases (ROCK1 and ROCK2). As would therefore be expected, lung ROCK activation has been demonstrated in humans with IPF and in animal models of this disease. ROCK inhibitors can prevent fibrosis in these models, and more importantly, induce the regression of already established fibrosis. Here we review ROCK structure and function, upstream activators and downstream targets of ROCKs in pulmonary fibrosis, contributions of ROCKs to profibrotic cellular responses to lung injury, ROCK inhibitors and their efficacy in animal models of pulmonary fibrosis, and potential toxicities of ROCK inhibitors in humans, as well as involvement of ROCKs in fibrosis in other organs. As we discuss, ROCK activation is required for multiple profibrotic responses, in the lung and multiple other organs, suggesting ROCK participation in fundamental pathways that contribute to the pathogenesis of a broad array of fibrotic diseases. Multiple lines of evidence therefore indicate that ROCK inhibition has great potential to be a powerful therapeutic tool in the treatment of fibrosis, both in the lung and beyond.
Collapse
Affiliation(s)
- Rachel S Knipe
- Pulmonary and Critical Care Unit and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (R.S.K., A.M.T.); and Section of Cardiology, Department of Medicine, University of Chicago, Chicago, Illinois (J.K.L.)
| | - Andrew M Tager
- Pulmonary and Critical Care Unit and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (R.S.K., A.M.T.); and Section of Cardiology, Department of Medicine, University of Chicago, Chicago, Illinois (J.K.L.)
| | - James K Liao
- Pulmonary and Critical Care Unit and Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts (R.S.K., A.M.T.); and Section of Cardiology, Department of Medicine, University of Chicago, Chicago, Illinois (J.K.L.)
| |
Collapse
|
15
|
Abstract
Lymphatic vessels (LVs) are involved in a number of physiological and pathophysiological processes such as fluid homoeostasis, immune surveillance, and resolution of inflammation and wound healing. Lymphangiogenesis, the outgrowth of existing LVs and the formation of new ones, has received increasing attention over the past decade on account of its prominence in organ physiology and pathology, which has been enabled by the development of specific tools to study lymph vessel functions. Several studies have been devoted to renal lymphatic vasculature and lymphangiogenesis in kidney diseases, such as chronic renal transplant dysfunction, primary renal fibrotic disorders, proteinuria, diabetic nephropathy and renal inflammation. This review describes the most recent findings on lymphangiogenesis, with a specific focus on renal lymphangiogenesis and its impact on renal diseases. We suggest renal lymphatics as a possible target for therapeutic interventions in renal medicine to dampen tubulointerstitial tissue remodelling and improve renal functioning.
Collapse
|
16
|
Jiang X, Tian W, Sung YK, Qian J, Nicolls MR. Macrophages in solid organ transplantation. Vasc Cell 2014; 6:5. [PMID: 24612731 PMCID: PMC3975229 DOI: 10.1186/2045-824x-6-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Accepted: 02/25/2014] [Indexed: 12/19/2022] Open
Abstract
Macrophages are highly plastic hematopoietic cells with diversified functions related to their anatomic location and differentiation states. A number of recent studies have examined the role of macrophages in solid organ transplantation. These studies show that macrophages can induce allograft injury but, conversely, can also promote tissue repair in ischemia-reperfusion injury and acute rejection. Therapeutic strategies that target macrophages to improve outcomes in solid organ transplant recipients are being examined in preclinical and clinical models. In this review, we discuss the role of macrophages in different types of injury and rejection, with a focus on macrophage-mediated tissue injury, specifically vascular injury, repair and remodeling. We also discuss emerging macrophage-centered therapeutic opportunities in solid organ transplantation.
Collapse
Affiliation(s)
- Xinguo Jiang
- Department of Medicine, VA Palo Alto Health Care System/Division of Pulmonary/Critical Care, Stanford University School of Medicine, Stanford, CA 94304, USA.
| | | | | | | | | |
Collapse
|