1
|
Katagiri D, Nagasaka S, Takahashi K, Wang S, Pozzi A, Zent R, Shimizu A, Zhang MZ, Göthert JR, van Kuppevelt TH, Harris RC, Takahashi T. Endothelial eNOS deficiency causes podocyte injury through NFAT2 and heparanase in diabetic mice. Sci Rep 2024; 14:29179. [PMID: 39587144 PMCID: PMC11589149 DOI: 10.1038/s41598-024-79501-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 11/11/2024] [Indexed: 11/27/2024] Open
Abstract
The pivotal role of endothelial nitric oxide synthase (eNOS) in diabetic nephropathy (DN) has been demonstrated using global eNOS knockout (eNOSGKO) mice. However, the precise role of endothelially expressed eNOS and how its deficiency advances DN are still unclear. Here, we targeted endothelial eNOS expression (E-eNOSKO) after the onset of diabetes using the floxed eNOS and endSCL-CreERT alleles. Diabetes was induced by low-dose streptozotocin injections. To evaluate the role of nuclear factor of activated T cells-2 (NFAT2) in podocyte injury in this condition, podocyte-specific NFAT2KO mice were also generated on eNOSGKO mice. The mechanisms of podocyte injury were investigated using cultured podocytes. Compared with diabetic wild-type mice, diabetic E-eNOSKO mice showed more advanced DN accompanied by NFAT2 expression in podocytes. NO donor suppressed NFAT2 expression and activation in high-glucose cultured podocytes as well as in diabetic E-eNOSKO mice. Furthermore, podocyte-specific deletion of NFAT2 attenuated DN in diabetic eNOSGKO mice accompanied by decreased heparanase (HPSE) expression in podocytes. Consistent with this finding, HPSE was upregulated by NFAT2 transfection and suppressed by NFAT2 siRNA or NO donor treatment in cultured podocytes. HPSE transfection reduced podocyte attachment to extracellular matrix concurrent with syndecan-4 (SDC4) shedding, and this effect was attenuated by co-transfection of SDC4. Finally, HPSE inhibitor treatment attenuated podocyte injury in diabetic E-eNOSKO mice with increased SDC4 expression in podocytes. Collectively, our data suggest that endothelial eNOS deficiency causes podocyte HPSE expression in diabetic mice through NFAT2, and HPSE promotes podocyte detachment in part through SDC4 shedding, advancing DN.
Collapse
Affiliation(s)
- Daisuke Katagiri
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
| | - Shinya Nagasaka
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Keiko Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
| | - Suwan Wang
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
| | - Ambra Pozzi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
- Department of Veterans Affairs, Nashville, TN, USA
| | - Roy Zent
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
- Department of Veterans Affairs, Nashville, TN, USA
| | - Akira Shimizu
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Ming-Zhi Zhang
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
| | - Joachim R Göthert
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center, University Hospital Essen, Essen, Germany
| | - Toin H van Kuppevelt
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Raymond C Harris
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA
- Department of Veterans Affairs, Nashville, TN, USA
| | - Takamune Takahashi
- Division of Nephrology and Hypertension, Vanderbilt University Medical Center, S-3223, MCN, Nashville, TN, 37232, USA.
| |
Collapse
|
2
|
Noda K, Atale N, Al‐Zahrani A, Furukawa M, Snyder ME, Ren X, Sanchez PG. Heparanase-induced endothelial glycocalyx degradation exacerbates lung ischemia/reperfusion injury in male mice. Physiol Rep 2024; 12:e70113. [PMID: 39448392 PMCID: PMC11502304 DOI: 10.14814/phy2.70113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
The endothelial glycocalyx (eGC) is a carbohydrate-rich layer on the vascular endothelium, and its damage can lead to endothelial and organ dysfunction. Heparanase (HPSE) degrades the eGC in response to cellular stress, but its role in organ dysfunction remains unclear. This study investigates HPSE's role in lung ischemia-reperfusion (I/R) injury. A left lung hilar occlusion model was used in B6 wildtype (WT) and HPSE genetic knockout (-/-) mice to induce I/R injury in vivo. The left lungs were ischemic for 1 h followed by reperfusion for 4 h prior to investigations of lung function and eGC status. Data were compared between uninjured lungs and I/R-injured lungs in WT and HPSE-/- mice. WT lungs showed significant functional impairment after I/R injury, whereas HPSE-/- lungs did not. Inhibition or knockout of HPSE prevented eGC damage, inflammation, and cellular migration after I/R injury by reducing matrix metalloproteinase activities. HPSE-/- mice exhibited compensatory regulation of related gene expressions. HPSE facilitates eGC degradation leading to inflammation and impaired lung function after I/R injury. HPSE may be a therapeutic target to attenuate graft damage in lung transplantation.
Collapse
Affiliation(s)
- Kentaro Noda
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Neha Atale
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Amer Al‐Zahrani
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Masashi Furukawa
- Division of Lung Transplant and Lung Failure, Department of Cardiothoracic SurgeryUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Mark E. Snyder
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Xi Ren
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Pablo G. Sanchez
- Section of Thoracic Surgery, Department of SurgeryUniversity of ChicagoChicagoIllinoisUSA
| |
Collapse
|
3
|
Vlodavsky I, Hilwi M, Kayal Y, Soboh S, Ilan N. Impact of heparanase-2 (Hpa2) on cancer and inflammation: Advances and paradigms. FASEB J 2024; 38:e23670. [PMID: 38747803 DOI: 10.1096/fj.202400286r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/09/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
HPSE2, the gene-encoding heparanase 2 (Hpa2), is mutated in urofacial syndrome (UFS), a rare autosomal recessive congenital disease attributed to peripheral neuropathy. Hpa2 lacks intrinsic heparan sulfate (HS)-degrading activity, the hallmark of heparanase (Hpa1), yet it exhibits a high affinity toward HS, thereby inhibiting Hpa1 enzymatic activity. Hpa2 regulates selected genes that promote normal differentiation, tissue homeostasis, and endoplasmic reticulum (ER) stress, resulting in antitumor, antiangiogenic, and anti-inflammatory effects. Importantly, stress conditions induce the expression of Hpa2, thus establishing a feedback loop, where Hpa2 enhances ER stress which, in turn, induces Hpa2 expression. In most cases, cancer patients who retain high levels of Hpa2 survive longer than patients bearing Hpa2-low tumors. Experimentally, overexpression of Hpa2 attenuates the growth of tumor xenografts, whereas Hpa2 gene silencing results in aggressive tumors. Studies applying conditional Hpa2 knockout (cHpa2-KO) mice revealed an essential involvement of Hpa2 contributed by the host in protecting against cancer and inflammation. This was best reflected by the distorted morphology of the Hpa2-null pancreas, including massive infiltration of immune cells, acinar to adipocyte trans-differentiation, and acinar to ductal metaplasia. Moreover, orthotopic inoculation of pancreatic ductal adenocarcinoma (PDAC) cells into the pancreas of Hpa2-null vs. wild-type mice yielded tumors that were by far more aggressive. Likewise, intravenous inoculation of cancer cells into cHpa2-KO mice resulted in a dramatically increased lung colonization reflecting the involvement of Hpa2 in restricting the formation of a premetastatic niche. Elucidating Hpa2 structure-activity-relationships is expected to support the development of Hpa2-based therapies against cancer and inflammation.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Maram Hilwi
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Yasmin Kayal
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Soaad Soboh
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Neta Ilan
- Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| |
Collapse
|
4
|
Peng QY, An Y, Jiang ZZ, Xu Y. The Role of Immune Cells in DKD: Mechanisms and Targeted Therapies. J Inflamm Res 2024; 17:2103-2118. [PMID: 38601771 PMCID: PMC11005934 DOI: 10.2147/jir.s457526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 03/19/2024] [Indexed: 04/12/2024] Open
Abstract
Diabetic kidney disease (DKD), is a common microvascular complication and a major cause of death in patients with diabetes. Disorders of immune cells and immune cytokines can accelerate DKD development of in a number of ways. As the kidney is composed of complex and highly differentiated cells, the interactions among different cell types and immune cells play important regulatory roles in disease development. Here, we summarize the latest research into the molecular mechanisms underlying the interactions among various immune and renal cells in DKD. In addition, we discuss the most recent studies related to single cell technology and bioinformatics analysis in the field of DKD. The aims of our review were to explore immune cells as potential therapeutic targets in DKD and provide some guidance for future clinical treatments.
Collapse
Affiliation(s)
- Qiu-Yue Peng
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, People’s Republic of China
| | - Ying An
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, People’s Republic of China
| | - Zong-Zhe Jiang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, People’s Republic of China
| | - Yong Xu
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, People’s Republic of China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Sichuan, People’s Republic of China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, Sichuan, People’s Republic of China
| |
Collapse
|
5
|
Gamez M, Elhegni HE, Fawaz S, Ho KH, Campbell NW, Copland DA, Onions KL, Butler MJ, Wasson EJ, Crompton M, Ramnath RD, Qiu Y, Yamaguchi Y, Arkill KP, Bates DO, Turnbull JE, Zubkova OV, Welsh GI, Atan D, Satchell SC, Foster RR. Heparanase inhibition as a systemic approach to protect the endothelial glycocalyx and prevent microvascular complications in diabetes. Cardiovasc Diabetol 2024; 23:50. [PMID: 38302978 PMCID: PMC10835837 DOI: 10.1186/s12933-024-02133-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Diabetes mellitus is a chronic disease which is detrimental to cardiovascular health, often leading to secondary microvascular complications, with huge global health implications. Therapeutic interventions that can be applied to multiple vascular beds are urgently needed. Diabetic retinopathy (DR) and diabetic kidney disease (DKD) are characterised by early microvascular permeability changes which, if left untreated, lead to visual impairment and renal failure, respectively. The heparan sulphate cleaving enzyme, heparanase, has previously been shown to contribute to diabetic microvascular complications, but the common underlying mechanism which results in microvascular dysfunction in conditions such as DR and DKD has not been determined. METHODS In this study, two mouse models of heparan sulphate depletion (enzymatic removal and genetic ablation by endothelial specific Exotosin-1 knock down) were utilized to investigate the impact of endothelial cell surface (i.e., endothelial glycocalyx) heparan sulphate loss on microvascular barrier function. Endothelial glycocalyx changes were measured using fluorescence microscopy or transmission electron microscopy. To measure the impact on barrier function, we used sodium fluorescein angiography in the eye and a glomerular albumin permeability assay in the kidney. A type 2 diabetic (T2D, db/db) mouse model was used to determine the therapeutic potential of preventing heparan sulphate damage using treatment with a novel heparanase inhibitor, OVZ/HS-1638. Endothelial glycocalyx changes were measured as above, and microvascular barrier function assessed by albumin extravasation in the eye and a glomerular permeability assay in the kidney. RESULTS In both models of heparan sulphate depletion, endothelial glycocalyx depth was reduced and retinal solute flux and glomerular albumin permeability was increased. T2D mice treated with OVZ/HS-1638 had improved endothelial glycocalyx measurements compared to vehicle treated T2D mice and were simultaneously protected from microvascular permeability changes associated with DR and DKD. CONCLUSION We demonstrate that endothelial glycocalyx heparan sulphate plays a common mechanistic role in microvascular barrier function in the eye and kidney. Protecting the endothelial glycocalyx damage in diabetes, using the novel heparanase inhibitor OVZ/HS-1638, effectively prevents microvascular permeability changes associated with DR and DKD, demonstrating a novel systemic approach to address diabetic microvascular complications.
Collapse
Affiliation(s)
- Monica Gamez
- Bristol Renal, Bristol Medical School, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom.
| | - Hesham E Elhegni
- Bristol Renal, Bristol Medical School, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Sarah Fawaz
- Bristol Renal, Bristol Medical School, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Kwan Ho Ho
- Department of Computer Science, Merchant Venturers Building, University of Bristol, Woodland Road, Bristol, BS8 1UB, United Kingdom
| | - Neill W Campbell
- Department of Computer Science, Merchant Venturers Building, University of Bristol, Woodland Road, Bristol, BS8 1UB, United Kingdom
| | - David A Copland
- Academic Unit of Ophthalmology, Translational Health Sciences, Bristol Medical School, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, United Kingdom
| | - Karen L Onions
- Bristol Renal, Bristol Medical School, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Matthew J Butler
- Bristol Renal, Bristol Medical School, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Elizabeth J Wasson
- Bristol Renal, Bristol Medical School, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Michael Crompton
- Bristol Renal, Bristol Medical School, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Raina D Ramnath
- Bristol Renal, Bristol Medical School, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Yan Qiu
- Bristol Renal, Bristol Medical School, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Yu Yamaguchi
- Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Kenton P Arkill
- School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2UH, United Kingdom
| | - David O Bates
- School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2UH, United Kingdom
| | - Jeremy E Turnbull
- Centre for Glycoscience, School of Life Sciences, Keele University, Staffordshire, ST5 5BG, United Kingdom
| | - Olga V Zubkova
- Ferrier Research Institute, Victoria University of Wellington, 69 Gracefield Rd, Lower Hutt, 5046, New Zealand
| | - Gavin I Welsh
- Bristol Renal, Bristol Medical School, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Denize Atan
- Academic Unit of Ophthalmology, Translational Health Sciences, Bristol Medical School, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, United Kingdom
- Bristol Eye Hospital, University Hospitals Bristol & Weston NHS Foundation Trust, Bristol, BS1 2LX, United Kingdom
| | - Simon C Satchell
- Bristol Renal, Bristol Medical School, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| | - Rebecca R Foster
- Bristol Renal, Bristol Medical School, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, United Kingdom
| |
Collapse
|
6
|
Song J, Zhang B, Zhang H, Cheng W, Liu P, Kang J. Quantitative Proteomics Combined with Network Pharmacology Analysis Unveils the Biological Basis of Schisandrin B in Treating Diabetic Nephropathy. Comb Chem High Throughput Screen 2024; 27:284-297. [PMID: 37151069 DOI: 10.2174/1386207326666230505111903] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/16/2023] [Accepted: 03/22/2023] [Indexed: 05/09/2023]
Abstract
BACKGROUND Diabetic nephropathy (DN) is a major complication of diabetes. Schisandrin B (Sch) is a natural pharmaceutical monomer that was shown to prevent kidney damage caused by diabetes and restore its function. However, there is still a lack of comprehensive and systematic understanding of the mechanism of Sch treatment in DN. OBJECTIVE We aim to provide a systematic overview of the mechanisms of Sch in multiple pathways to treat DN in rats. METHODS Streptozocin was used to build a DN rat model, which was further treated with Sch. The possible mechanism of Sch protective effects against DN was predicted using network pharmacology and was verified by quantitative proteomics analysis. RESULTS High dose Sch treatment significantly downregulated fasting blood glucose, creatinine, blood urea nitrogen, and urinary protein levels and reduced collagen deposition in the glomeruli and tubule-interstitium of DN rats. The activities of superoxide dismutase (SOD) and plasma glutathione peroxidase (GSH-Px) in the kidney of DN rats significantly increased with Sch treatment. In addition, the levels of IL-6, IL-1β, and TNF-α were significantly reduced in DN rats treated with Sch. 11 proteins that target both Sch and DN were enriched in pathways such as MAPK signaling, PI3K-Akt signaling, renal cell carcinoma, gap junction, endocrine resistance, and TNF signaling. Furthermore, quantitative proteomics showed that Xaf1 was downregulated in the model vs. control group and upregulated in the Sch-treated vs. model group. Five proteins, Crb3, Tspan4, Wdr45, Zfp512, and Tmigd1, were found to be upregulated in the model vs. control group and downregulated in the Sch vs. model group. Three intersected proteins between the network pharmacology prediction and proteomics results, Crb3, Xaf1, and Tspan4, were identified. CONCLUSION Sch functions by relieving oxidative stress and the inflammatory response by regulating Crb3, Xaf1, and Tspan4 protein expression levels to treat DN disease.
Collapse
Affiliation(s)
- Jianying Song
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, People's Republic of China
| | - Bo Zhang
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, Beijing, 100084, China
| | - Huiping Zhang
- Shanghai Applied Protein Technology Co., Ltd., 58 Yuanmei Road, Shanghai, 200233, People's Republic of China
| | - Wenbo Cheng
- Tianjin Key Laboratory of Medical Mass Spectrometry for Accurate Diagnosis, Tianjin, 300399, People's Republic of China
| | - Peiyuan Liu
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, People's Republic of China
| | - Jun Kang
- School of Life Sciences, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, People's Republic of China
| |
Collapse
|
7
|
Locatelli M, Rottoli D, Mahmoud R, Abbate M, Corna D, Cerullo D, Tomasoni S, Remuzzi G, Zoja C, Benigni A, Macconi D. Endothelial Glycocalyx of Peritubular Capillaries in Experimental Diabetic Nephropathy: A Target of ACE Inhibitor-Induced Kidney Microvascular Protection. Int J Mol Sci 2023; 24:16543. [PMID: 38003732 PMCID: PMC10671403 DOI: 10.3390/ijms242216543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
Peritubular capillary rarefaction is a recurrent aspect of progressive nephropathies. We previously found that peritubular capillary density was reduced in BTBR ob/ob mice with type 2 diabetic nephropathy. In this model, we searched for abnormalities in the ultrastructure of peritubular capillaries, with a specific focus on the endothelial glycocalyx, and evaluated the impact of treatment with an angiotensin-converting enzyme inhibitor (ACEi). Mice were intracardially perfused with lanthanum to visualise the glycocalyx. Transmission electron microscopy analysis revealed endothelial cell abnormalities and basement membrane thickening in the peritubular capillaries of BTBR ob/ob mice compared to wild-type mice. Remodelling and focal loss of glycocalyx was observed in lanthanum-stained diabetic kidneys, associated with a reduction in glycocalyx components, including sialic acids, as detected through specific lectins. ACEi treatment preserved the endothelial glycocalyx and attenuated the ultrastructural abnormalities of peritubular capillaries. In diabetic mice, peritubular capillary damage was associated with an enhanced tubular expression of heparanase, which degrades heparan sulfate residues of the glycocalyx. Heparanase was also detected in renal interstitial macrophages that expressed tumor necrosis factor-α. All these abnormalities were mitigated by ACEi. Our findings suggest that, in experimental diabetic nephropathy, preserving the endothelial glycocalyx is important in order to protect peritubular capillaries from damage and loss.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, 24126 Bergamo, Italy; (M.L.); (D.R.); (R.M.); (M.A.); (D.C.); (D.C.); (S.T.); (G.R.); (C.Z.); (D.M.)
| | | |
Collapse
|
8
|
Li JC, Wang LJ, Feng F, Chen TT, Shi WG, Liu LP. Role of heparanase in sepsis‑related acute kidney injury (Review). Exp Ther Med 2023; 26:379. [PMID: 37456170 PMCID: PMC10347300 DOI: 10.3892/etm.2023.12078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 06/08/2023] [Indexed: 07/18/2023] Open
Abstract
Sepsis-related acute kidney injury (S-AKI) is a common and significant complication of sepsis in critically ill patients, which can often only be treated with antibiotics and medications that reduce S-AKI symptoms. The precise mechanism underlying the onset of S-AKI is still unclear, thus hindering the development of new strategies for its treatment. Therefore, it is necessary to explore the pathogenesis of S-AKI to identify biomarkers and therapeutic targets for its early diagnosis and treatment. Heparanase (HPA), the only known enzyme that cleaves the side chain of heparan sulfate, has been widely studied in relation to tumor metabolism, procoagulant activity, angiogenesis, inflammation and sepsis. It has been reported that HPA plays an important role in the progression of S-AKI. The aim of the present review was to provide an overview of the function of HPA in S-AKI and to summarize its underlying molecular mechanisms, including mediating inflammatory response, immune response, autophagy and exosome biogenesis. It is anticipated that emerging discoveries about HPA in S-AKI will support HPA as a potential biomarker and therapeutic target to combat S-AKI.
Collapse
Affiliation(s)
- Jian-Chun Li
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Lin-Jun Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Fei Feng
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Ting-Ting Chen
- The First Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Wen-Gui Shi
- Cuiying Biomedical Research Center, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Li-Ping Liu
- Department of Emergency, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
9
|
Buijsers B, Garsen M, de Graaf M, Bakker-van Bebber M, Guo C, Li X, van der Vlag J. Heparanase-2 protein and peptides have a protective effect on experimental glomerulonephritis and diabetic nephropathy. Front Pharmacol 2023; 14:1098184. [PMID: 37180718 PMCID: PMC10172501 DOI: 10.3389/fphar.2023.1098184] [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: 11/22/2022] [Accepted: 04/17/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction: The endothelial glycocalyx degrading enzyme heparanase-1 (HPSE1) is a major contributor to kidney diseases, such as glomerulonephritis and diabetic nephropathy. Therefore, inhibition of HPSE1 could be an interesting therapeutic strategy to treat glomerular diseases. A possible HPSE1 inhibitor is heparanase-2 (HPSE2) because HPSE2 is a structural homolog of HPSE1 without enzymatic activity. The importance of HPSE2 has been recently demonstrated in HPSE2-deficient mice, since these mice developed albuminuria and died within a few months after birth. We postulate that inhibition of HPSE1 activity by HPSE2 is a promising therapeutic strategy to target albuminuria and resulting renal failure. Methods: First, we evaluated the regulation of HPSE2 expression in anti-GBM and LPS-induced glomerulonephritis, streptozotocin-induced diabetic nephropathy, and adriamycin nephropathy by qPCR and ELISA. Second, we measured the HPSE1 inhibiting capacity of HPSE2 protein and 30 different HPSE2 peptides and assessed their therapeutic potential in both experimental glomerulonephritis and diabetic nephropathy using kidney function and cortical mRNA expression of HPSE1 and cytokines as outcome parameters. Results: HPSE2 expression was downregulated under inflammatory and diabetic conditions, whereas this effect on HPSE2 expression was absent with HPSE1 inhibition and in HPSE1-deficient mice. Both HPSE2 protein and a mixture of the three most potent HPSE1 inhibitory HPSE2 peptides could prevent LPS and streptozotocin induced kidney injury. Discussion: Taken together, our data suggest a protective effect of HPSE2 in (experimental) glomerular diseases and support the therapeutic potential of HPSE2 as HPSE1 inhibitor in glomerular diseases.
Collapse
Affiliation(s)
- Baranca Buijsers
- Department of Nephrology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Marjolein Garsen
- Department of Nephrology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mark de Graaf
- Department of Nephrology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Marinka Bakker-van Bebber
- Department of Nephrology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Chunming Guo
- Departments of Urology and Pathology, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Xue Li
- Departments of Urology and Pathology, Boston Children’s Hospital, Boston, MA, United States
- Department of Surgery, Harvard Medical School, Boston, MA, United States
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
10
|
van der Velden AIM, van den Berg BM, van den Born BJ, Galenkamp H, Ijpelaar DHT, Rabelink TJ. Ethnic differences in urinary monocyte chemoattractant protein-1 and heparanase-1 levels in individuals with type 2 diabetes: the HELIUS study. BMJ Open Diabetes Res Care 2022; 10:10/6/e003003. [PMID: 36564084 PMCID: PMC9791388 DOI: 10.1136/bmjdrc-2022-003003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/05/2022] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION We aimed to investigate ethnic differences in two urinary inflammatory markers in participants with type 2 diabetes mellitus (T2DM). RESEARCH DESIGN AND METHODS We included 55 Dutch, 127 South-Asian Surinamese, 92 African Surinamese, 62 Ghanaian, 74 Turkish and 88 Moroccan origin participants with T2DM from the HEalthy LIfe in an Urban Setting study. Using linear regression analyses, we investigated differences in urinary monocyte chemoattractant protein-1 (MCP-1) and heparanase-1 (HPSE-1) levels across ethnic minorities compared with Dutch. Associations between the urinary markers and albuminuria (albumin:creatinine ratio (ACR)) was investigated per ethnicity. RESULTS Urinary MCP-1 levels were higher in the Moroccan participants (0.15 log ng/mmol, 95% CI 0.05 to 0.26) compared with Dutch after multiple adjustments. Urinary HPSE-1 levels were lower in the African Surinamese and Ghanaian participants compared with the Dutch, with a difference of -0.16 log mU/mmol (95% CI -0.29 to -0.02) in African Surinamese and -0.16 log mU/mmol (95% CI -0.31 to -0.00) in Ghanaian after multiple adjustments. In all ethnic groups except the Dutch and Ghanaian participants, MCP-1 was associated with ACR. This association remained strongest after multiple adjustment in South-Asian and African Surinamese participants, with an increase in log ACR of 1.03% (95% CI 0.58 to 1.47) and 1.23% (95% CI 0.52 to 1.94) if log MCP-1 increased 1%. Only in the Dutch participants, an association between HPSE-1 and ACR was found, with increase in log ACR of 0.40% (95% CI 0.04 to 0.76) if log HPSE-1 increased 1%. CONCLUSIONS We found ethnic differences in urinary MCP-1 and HPSE-1 levels, in a multi-ethnic cohort of participants with T2DM. In addition, we found ethnic differences in the association of MCP-1 and HPSE-1 levels with albuminuria. These findings suggest differences in renal inflammation across ethnic groups.
Collapse
Affiliation(s)
- Anouk I M van der Velden
- Department of Internal Medicine (Nephrology) and Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Bernard M van den Berg
- Department of Internal Medicine (Nephrology) and Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - B J van den Born
- Internal Medicine, Amsterdam UMC-Locatie AMC, Amsterdam, The Netherlands
| | - Henrike Galenkamp
- Public and Occupational Health, Amsterdam UMC-Locatie AMC, Amsterdam, The Netherlands
| | - Daphne H T Ijpelaar
- Department of Internal Medicine (Nephrology) and Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
- Department of Internal Medicine and Nephrology, Groene Hart Hospital, Gouda, The Netherlands
| | - Ton J Rabelink
- Department of Internal Medicine (Nephrology) and Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
11
|
Heparanase Increases Podocyte Survival and Autophagic Flux after Adriamycin-Induced Injury. Int J Mol Sci 2022; 23:ijms232012691. [PMID: 36293542 PMCID: PMC9604275 DOI: 10.3390/ijms232012691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/06/2022] [Accepted: 10/19/2022] [Indexed: 11/17/2022] Open
Abstract
The kidney glomerular filtration barrier (GFB) is enriched with heparan sulfate (HS) proteoglycans, which contribute to its permselectivity. The endoglycosidase heparanase cleaves HS and hence appears to be involved in the pathogenesis of kidney injury and glomerulonephritis. We have recently reported, nonetheless, that heparanase overexpression preserved glomerular structure and kidney function in an experimental model of Adriamycin-induced nephropathy. To elucidate mechanisms underlying heparanase function in podocytes-key GFB cells, we utilized a human podocyte cell line and transgenic mice overexpressing heparanase. Notably, podocytes overexpressing heparanase (H) demonstrated significantly higher survival rates and viability after exposure to Adriamycin or hydrogen peroxide, compared with mock-infected (V) podocytes. Immunofluorescence staining of kidney cryo-sections and cultured H and V podocytes as well as immunoblotting of proteins extracted from cultured cells, revealed that exposure to toxic injury resulted in a significant increase in autophagic flux in H podocytes, which was reversed by the heparanase inhibitor, Roneparstat (SST0001). Heparanase overexpression was also associated with substantial transcriptional upregulation of autophagy genes BCN1, ATG5, and ATG12, following Adriamycin treatment. Moreover, cleaved caspase-3 was attenuated in H podocytes exposed to Adriamycin, indicating lower apoptotic cell death in H vs. V podocytes. Collectively, these findings suggest that in podocytes, elevated levels of heparanase promote cytoprotection.
Collapse
|
12
|
Heparanase: A Novel Therapeutic Target for the Treatment of Atherosclerosis. Cells 2022; 11:cells11203198. [PMID: 36291066 PMCID: PMC9599978 DOI: 10.3390/cells11203198] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death and disability worldwide, and its management places a huge burden on healthcare systems through hospitalisation and treatment. Atherosclerosis is a chronic inflammatory disease of the arterial wall resulting in the formation of lipid-rich, fibrotic plaques under the subendothelium and is a key contributor to the development of CVD. As such, a detailed understanding of the mechanisms involved in the development of atherosclerosis is urgently required for more effective disease treatment and prevention strategies. Heparanase is the only mammalian enzyme known to cleave heparan sulfate of heparan sulfate proteoglycans, which is a key component of the extracellular matrix and basement membrane. By cleaving heparan sulfate, heparanase contributes to the regulation of numerous physiological and pathological processes such as wound healing, inflammation, tumour angiogenesis, and cell migration. Recent evidence suggests a multifactorial role for heparanase in atherosclerosis by promoting underlying inflammatory processes giving rise to plaque formation, as well as regulating lesion stability. This review provides an up-to-date overview of the role of heparanase in physiological and pathological processes with a focus on the emerging role of the enzyme in atherosclerosis.
Collapse
|
13
|
Macrophages Upregulate Estrogen Receptor Expression in the Model of Obesity-Associated Breast Carcinoma. Cells 2022; 11:cells11182844. [PMID: 36139419 PMCID: PMC9496942 DOI: 10.3390/cells11182844] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
Breast cancer (BC) and obesity are two heterogeneous conditions with a tremendous impact on health. BC is the most commonly diagnosed neoplasm and the leading cause of cancer-related mortality among women, and the prevalence of obesity in women worldwide reaches pandemic proportions. Obesity is a significant risk factor for both incidence and worse prognosis in estrogen receptor positive (ER+) BC. Yet, the mechanisms underlying the association between excess adiposity and increased risk/therapy resistance/poorer outcome of ER+, but not ER−negative (ER−), BC are not fully understood. Tumor-promoting action of obesity, predominantly in ER + BC patients, is often attributed to the augmented production of estrogen in ‘obese’ adipose tissue. However, in addition to the estrogen production, expression levels of ER represent a key determinant in hormone-driven breast tumorigenesis and therapy response. Here, utilizing in vitro and in vivo models of BC, we show that macrophages, whose adverse activation by obesogenic substances is fueled by heparanase (extracellular matrix-degrading enzyme), are capable of upregulating ER expression in tumor cells, in the setting of obesity-associated BC. These findings underscore a previously unknown mechanism through which interplay between cellular/extracellular elements of obesity-associated BC microenvironment influences estrogen sensitivity—a critical component in hormone-related cancer progression and resistance to therapy.
Collapse
|
14
|
Foote CA, Soares RN, Ramirez-Perez FI, Ghiarone T, Aroor A, Manrique-Acevedo C, Padilla J, Martinez-Lemus LA. Endothelial Glycocalyx. Compr Physiol 2022; 12:3781-3811. [PMID: 35997082 PMCID: PMC10214841 DOI: 10.1002/cphy.c210029] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The glycocalyx is a polysaccharide structure that protrudes from the body of a cell. It is primarily conformed of glycoproteins and proteoglycans, which provide communication, electrostatic charge, ionic buffering, permeability, and mechanosensation-mechanotransduction capabilities to cells. In blood vessels, the endothelial glycocalyx that projects into the vascular lumen separates the vascular wall from the circulating blood. Such a physical location allows a number of its components, including sialic acid, glypican-1, heparan sulfate, and hyaluronan, to participate in the mechanosensation-mechanotransduction of blood flow-dependent shear stress, which results in the synthesis of nitric oxide and flow-mediated vasodilation. The endothelial glycocalyx also participates in the regulation of vascular permeability and the modulation of inflammatory responses, including the processes of leukocyte rolling and extravasation. Its structural architecture and negative charge work to prevent macromolecules greater than approximately 70 kDa and cationic molecules from binding and flowing out of the vasculature. This also prevents the extravasation of pathogens such as bacteria and virus, as well as that of tumor cells. Due to its constant exposure to shear and circulating enzymes such as neuraminidase, heparanase, hyaluronidase, and matrix metalloproteinases, the endothelial glycocalyx is in a continuous process of degradation and renovation. A balance favoring degradation is associated with a variety of pathologies including atherosclerosis, hypertension, vascular aging, metastatic cancer, and diabetic vasculopathies. Consequently, ongoing research efforts are focused on deciphering the mechanisms that promote glycocalyx degradation or limit its syntheses, as well as on therapeutic approaches to improve glycocalyx integrity with the goal of reducing vascular disease. © 2022 American Physiological Society. Compr Physiol 12: 1-31, 2022.
Collapse
Affiliation(s)
- Christopher A. Foote
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Rogerio N. Soares
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | | | - Thaysa Ghiarone
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
| | - Annayya Aroor
- Department of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
| | - Camila Manrique-Acevedo
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medicine, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
| | - Jaume Padilla
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, USA
| | - Luis A. Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, USA
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
15
|
Liu T, Li H, Conley YP, Primack BA, Wang J, Lo WJ, Li C. A Genome-Wide Association Study of Prediabetes Status Change. Front Endocrinol (Lausanne) 2022; 13:881633. [PMID: 35769078 PMCID: PMC9234217 DOI: 10.3389/fendo.2022.881633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
We conducted the first genome-wide association study of prediabetes status change (to diabetes or normal glycaemia) among 900 White participants of the Atherosclerosis Risk in Communities (ARIC) study. Single nucleotide polymorphism (SNP)-based analysis was performed by logistic regression models, controlling for age, gender, body mass index, and the first 3 genetic principal components. Gene-based analysis was conducted by combining SNP-based p values using effective Chi-square test method. Promising SNPs (p < 1×10-5) and genes (p < 1×10-4) were further evaluated for replication among 514 White participants of the Framingham Heart Study (FHS). To accommodate familial correlations, generalized estimation equation models were applied for SNP-based analyses in the FHS. Analysis results across ARIC and FHS were combined using inverse-variance-weighted meta-analysis method for SNPs and Fisher's method for genes. We robustly identified 5 novel genes that are associated with prediabetes status change using gene-based analyses, including SGCZ (ARIC p = 9.93×10-6, FHS p = 2.00×10-3, Meta p = 3.72×10-7) at 8p22, HPSE2 (ARIC p = 8.26×10-19, FHS p = 5.85×10-3, Meta p < 8.26×10-19) at 10q24.2, ADGRA1 (ARIC p = 1.34×10-5, FHS p = 1.13×10-3, Meta p = 2.88×10-7) at 10q26.3, GLB1L3 (ARIC p = 3.71×10-6, FHS p = 4.51×10-3, Meta p = 3.16×10-7) at 11q25, and PCSK6 (ARIC p = 6.51×10-6, FHS p = 1.10×10-2, Meta p = 1.25×10-6) at 15q26.3. eQTL analysis indicated that these genes were highly expressed in tissues related to diabetes development. However, we were not able to identify any novel locus in single SNP-based analysis. Future large scale genomic studies of prediabetes status change are warranted.
Collapse
Affiliation(s)
- Tingting Liu
- College of Nursing, Florida State University, Tallahassee, FL, United States
| | - Hongjin Li
- College of Nursing, University of Illinois at Chicago, Chicago, IL, United States
| | - Yvette P. Conley
- School of Nursing, University of Pittsburgh, Pittsburgh, PA, United States
| | - Brian A. Primack
- College of Education and Health Professions, University of Arkansas, Fayetteville, AR, United States
| | - Jing Wang
- College of Nursing, Florida State University, Tallahassee, FL, United States
| | - Wen-Juo Lo
- College of Education and Health Professions, University of Arkansas, Fayetteville, AR, United States
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Tropical Medicine and Public Health, New Orleans, LA, United States
- *Correspondence: Changwei Li,
| |
Collapse
|
16
|
Vlodavsky I, Barash U, Nguyen HM, Yang SM, Ilan N. Biology of the Heparanase-Heparan Sulfate Axis and Its Role in Disease Pathogenesis. Semin Thromb Hemost 2021; 47:240-253. [PMID: 33794549 DOI: 10.1055/s-0041-1725066] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell surface proteoglycans are important constituents of the glycocalyx and participate in cell-cell and cell-extracellular matrix (ECM) interactions, enzyme activation and inhibition, and multiple signaling routes, thereby regulating cell proliferation, survival, adhesion, migration, and differentiation. Heparanase, the sole mammalian heparan sulfate degrading endoglycosidase, acts as an "activator" of HS proteoglycans, thus regulating tissue hemostasis. Heparanase is a multifaceted enzyme that together with heparan sulfate, primarily syndecan-1, drives signal transduction, immune cell activation, exosome formation, autophagy, and gene transcription via enzymatic and nonenzymatic activities. An important feature is the ability of heparanase to stimulate syndecan-1 shedding, thereby impacting cell behavior both locally and distally from its cell of origin. Heparanase releases a myriad of HS-bound growth factors, cytokines, and chemokines that are sequestered by heparan sulfate in the glycocalyx and ECM. Collectively, the heparan sulfate-heparanase axis plays pivotal roles in creating a permissive environment for cell proliferation, differentiation, and function, often resulting in the pathogenesis of diseases such as cancer, inflammation, endotheliitis, kidney dysfunction, tissue fibrosis, and viral infection.
Collapse
Affiliation(s)
- Israel Vlodavsky
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Uri Barash
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Hien M Nguyen
- Department of Chemistry, Wayne State University, Detroit, Michigan
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, People's Republic of China
| | - Neta Ilan
- Technion Integrated Cancer Center (TICC), Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
17
|
Hermano E, Carlotti F, Abecassis A, Meirovitz A, Rubinstein AM, Li JP, Vlodavsky I, Rabelink TJ, Elkin M. Dichotomic role of heparanase in a murine model of metabolic syndrome. Cell Mol Life Sci 2021; 78:2771-2780. [PMID: 33051777 PMCID: PMC11072560 DOI: 10.1007/s00018-020-03660-2] [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: 05/13/2020] [Revised: 09/17/2020] [Accepted: 09/28/2020] [Indexed: 02/07/2023]
Abstract
Heparanase is the predominant enzyme that cleaves heparan sulfate, the main polysaccharide in the extracellular matrix. While the role of heparanase in sustaining the pathology of autoimmune diabetes is well documented, its association with metabolic syndrome/type 2 diabetes attracted less attention. Our research was undertaken to elucidate the significance of heparanase in impaired glucose metabolism in metabolic syndrome and early type 2 diabetes. Here, we report that heparanase exerts opposite effects in insulin-producing (i.e., islets) vs. insulin-target (i.e., skeletal muscle) compartments, sustaining or hampering proper regulation of glucose homeostasis depending on the site of action. We observed that the enzyme promotes macrophage infiltration into islets in a murine model of metabolic syndrome, and fosters β-cell-damaging properties of macrophages activated in vitro by components of diabetogenic/obese milieu (i.e., fatty acids). On the other hand, in skeletal muscle (prototypic insulin-target tissue), heparanase is essential to ensure insulin sensitivity. Thus, despite a deleterious effect of heparanase on macrophage infiltration in islets, the enzyme appears to have beneficial role in glucose homeostasis in metabolic syndrome. The dichotomic action of the enzyme in the maintenance of glycemic control should be taken into account when considering heparanase-targeting strategies for the treatment of diabetes.
Collapse
Affiliation(s)
- Esther Hermano
- Department of Oncology, Sharett Institute, Hadassah-Hebrew University Medical Center, 91120, Jerusalem, Israel
| | - Françoise Carlotti
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Alexia Abecassis
- Department of Oncology, Sharett Institute, Hadassah-Hebrew University Medical Center, 91120, Jerusalem, Israel
| | - Amichay Meirovitz
- Department of Oncology, Sharett Institute, Hadassah-Hebrew University Medical Center, 91120, Jerusalem, Israel
| | - Ariel M Rubinstein
- Department of Oncology, Sharett Institute, Hadassah-Hebrew University Medical Center, 91120, Jerusalem, Israel
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research Center, The Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Ton J Rabelink
- Division of Nephrology, Department of Internal Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Michael Elkin
- Department of Oncology, Sharett Institute, Hadassah-Hebrew University Medical Center, 91120, Jerusalem, Israel.
- Hebrew University Medical School, 91120, Jerusalem, Israel.
| |
Collapse
|
18
|
Rangarajan S, Richter JR, Richter RP, Bandari SK, Tripathi K, Vlodavsky I, Sanderson RD. Heparanase-enhanced Shedding of Syndecan-1 and Its Role in Driving Disease Pathogenesis and Progression. J Histochem Cytochem 2020; 68:823-840. [PMID: 32623935 PMCID: PMC7711244 DOI: 10.1369/0022155420937087] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 05/29/2020] [Indexed: 02/08/2023] Open
Abstract
Both heparanase and syndecan-1 are known to be present and active in disease pathobiology. An important feature of syndecan-1 related to its role in pathologies is that it can be shed from the surface of cells as an intact ectodomain composed of the extracellular core protein and attached heparan sulfate and chondroitin sulfate chains. Shed syndecan-1 remains functional and impacts cell behavior both locally and distally from its cell of origin. Shedding of syndecan-1 is initiated by a variety of stimuli and accomplished predominantly by the action of matrix metalloproteinases. The accessibility of these proteases to the core protein of syndecan-1 is enhanced, and shedding facilitated, when the heparan sulfate chains of syndecan-1 have been shortened by the enzymatic activity of heparanase. Interestingly, heparanase also enhances shedding by upregulating the expression of matrix metalloproteinases. Recent studies have revealed that heparanase-induced syndecan-1 shedding contributes to the pathogenesis and progression of cancer and viral infection, as well as other septic and non-septic inflammatory states. This review discusses the heparanase/shed syndecan-1 axis in disease pathogenesis and progression, the potential of targeting this axis therapeutically, and the possibility that this axis is widespread and of influence in many diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Israel Vlodavsky
- The University of Alabama at Birmingham, Birmingham, Alabama, and Technion Integrated Cancer Center, Rappaport Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | | |
Collapse
|
19
|
Li Z, Wu N, Wang J, Zhang Q. Roles of Endovascular Calyx Related Enzymes in Endothelial Dysfunction and Diabetic Vascular Complications. Front Pharmacol 2020; 11:590614. [PMID: 33328998 PMCID: PMC7734331 DOI: 10.3389/fphar.2020.590614] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 10/16/2020] [Indexed: 12/25/2022] Open
Abstract
In recent years, the number of diabetic patients has rapidly increased. Diabetic vascular complications seriously affect people’s quality of life. Studies found that endothelial dysfunction precedes the vascular complications of diabetes. Endothelial dysfunction is related to glycocalyx degradation on the surface of blood vessels. Heparanase (HPSE), matrix metalloproteinase (MMP), hyaluronidase (HYAL), hyaluronic acid synthase (HAS), and neuraminidase (NEU) are related to glycocalyx degradation. Therefore, we reviewed the relationship between endothelial dysfunction and the vascular complications of diabetes from the perspective of enzymes.
Collapse
Affiliation(s)
- Zhi Li
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Science and Technology, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ning Wu
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China.,Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jing Wang
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Science and Technology, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Quanbin Zhang
- Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China.,Lab for Marine Biology and Biotechnology, Qingdao National Lab for Marine Science and Technology, Qingdao, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
20
|
Abstract
Heparanase is the only mammalian enzyme that cleaves heparan sulphate, an important component of the extracellular matrix. This leads to the remodelling of the extracellular matrix, whilst liberating growth factors and cytokines bound to heparan sulphate. This in turn promotes both physiological and pathological processes such as angiogenesis, immune cell migration, inflammation, wound healing and metastasis. Furthermore, heparanase exhibits non-enzymatic actions in cell signalling and in regulating gene expression. Cancer is underpinned by key characteristic features that promote malignant growth and disease progression, collectively termed the 'hallmarks of cancer'. Essentially, all cancers examined to date have been reported to overexpress heparanase, leading to enhanced tumour growth and metastasis with concomitant poor patient survival. With its multiple roles within the tumour microenvironment, heparanase has been demonstrated to regulate each of these hallmark features, in turn highlighting the need for heparanase-targeted therapies. However, recent discoveries which demonstrated that heparanase can also regulate vital anti-tumour mechanisms have cast doubt on this approach. This review will explore the myriad ways by which heparanase functions as a key regulator of the hallmarks of cancer and will highlight its role as a major component within the tumour microenvironment. The dual role of heparanase within the tumour microenvironment, however, emphasises the need for further investigation into defining its precise mechanism of action in different cancer settings.
Collapse
Affiliation(s)
- Krishnath M Jayatilleke
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Plenty Road & Kingsbury Drive, Melbourne, VIC, 3086, Australia
| | - Mark D Hulett
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Plenty Road & Kingsbury Drive, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
21
|
Buijsers B, Yanginlar C, de Nooijer A, Grondman I, Maciej-Hulme ML, Jonkman I, Janssen NAF, Rother N, de Graaf M, Pickkers P, Kox M, Joosten LAB, Nijenhuis T, Netea MG, Hilbrands L, van de Veerdonk FL, Duivenvoorden R, de Mast Q, van der Vlag J. Increased Plasma Heparanase Activity in COVID-19 Patients. Front Immunol 2020; 11:575047. [PMID: 33123154 PMCID: PMC7573491 DOI: 10.3389/fimmu.2020.575047] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/14/2020] [Indexed: 12/23/2022] Open
Abstract
Reports suggest a role of endothelial dysfunction and loss of endothelial barrier function in COVID-19. It is well established that the endothelial glycocalyx-degrading enzyme heparanase contributes to vascular leakage and inflammation. Low molecular weight heparins (LMWH) serve as an inhibitor of heparanase. We hypothesize that heparanase contributes to the pathogenesis of COVID-19, and that heparanase may be inhibited by LMWH. To test this hypothesis, heparanase activity and heparan sulfate levels were measured in plasma of healthy controls (n = 10) and COVID-19 patients (n = 48). Plasma heparanase activity and heparan sulfate levels were significantly elevated in COVID-19 patients. Heparanase activity was associated with disease severity including the need for intensive care, lactate dehydrogenase levels, and creatinine levels. Use of prophylactic LMWH in non-ICU patients was associated with a reduced heparanase activity. Since there is no other clinically applied heparanase inhibitor currently available, therapeutic treatment of COVID-19 patients with low molecular weight heparins should be explored.
Collapse
Affiliation(s)
- Baranca Buijsers
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Cansu Yanginlar
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Aline de Nooijer
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Inge Grondman
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Marissa L. Maciej-Hulme
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Inge Jonkman
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nico A. F. Janssen
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nils Rother
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mark de Graaf
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Peter Pickkers
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Matthijs Kox
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Leo A. B. Joosten
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Tom Nijenhuis
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mihai G. Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
- Deparment of Immunology and Metabolism, Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Luuk Hilbrands
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Frank L. van de Veerdonk
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Raphaël Duivenvoorden
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Quirijn de Mast
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
22
|
Buijsers B, Yanginlar C, Maciej-Hulme ML, de Mast Q, van der Vlag J. Beneficial non-anticoagulant mechanisms underlying heparin treatment of COVID-19 patients. EBioMedicine 2020; 59:102969. [PMID: 32853989 PMCID: PMC7445140 DOI: 10.1016/j.ebiom.2020.102969] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/05/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease-2019 (COVID-19) is associated with severe inflammation in mainly the lung, and kidney. Reports suggest a beneficial effect of the use of heparin/low molecular weight heparin (LMWH) on mortality in COVID-19. In part, this beneficial effect could be explained by the anticoagulant properties of heparin/LMWH. Here, we summarise potential beneficial, non-anticoagulant mechanisms underlying treatment of COVID-19 patients with heparin/LMWH, which include: (i) Inhibition of heparanase activity, responsible for endothelial leakage; (ii) Neutralisation of chemokines, and cytokines; (iii) Interference with leukocyte trafficking; (iv) Reducing viral cellular entry, and (v) Neutralisation of extracellular cytotoxic histones. Considering the multiple inflammatory and pathogenic mechanisms targeted by heparin/LMWH, it is warranted to conduct clinical studies that evaluate therapeutic doses of heparin/LMWH in COVID-19 patients. In addition, identification of specific heparin-derived sequences that are functional in targeting non-anticoagulant mechanisms may have even higher therapeutic potential for COVID-19 patients, and patients suffering from other inflammatory diseases.
Collapse
Affiliation(s)
- Baranca Buijsers
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Cansu Yanginlar
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Marissa L Maciej-Hulme
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Quirijn de Mast
- Department of Internal Medicine, Radboud university medical center, Nijmegen, The Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
23
|
Pessentheiner AR, Ducasa GM, Gordts PLSM. Proteoglycans in Obesity-Associated Metabolic Dysfunction and Meta-Inflammation. Front Immunol 2020; 11:769. [PMID: 32508807 PMCID: PMC7248225 DOI: 10.3389/fimmu.2020.00769] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022] Open
Abstract
Proteoglycans are a specific subset of glycoproteins found at the cell surface and in the extracellular matrix, where they interact with a plethora of proteins involved in metabolic homeostasis and meta-inflammation. Over the last decade, new insights have emerged on the mechanism and biological significance of these interactions in the context of diet-induced disorders such as obesity and type-2 diabetes. Complications of energy metabolism drive most diet-induced metabolic disorders, which results in low-grade chronic inflammation, thereby affecting proper function of many vital organs involved in energy homeostasis, such as the brain, liver, kidney, heart and adipose tissue. Here, we discuss how heparan, chondroitin and keratan sulfate proteoglycans modulate obesity-induced metabolic dysfunction and low-grade inflammation that impact the initiation and progression of obesity-associated morbidities.
Collapse
Affiliation(s)
- Ariane R. Pessentheiner
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, United States
| | - G. Michelle Ducasa
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, United States
| | - Philip L. S. M. Gordts
- Department of Medicine, Division of Endocrinology and Metabolism, University of California, San Diego, La Jolla, CA, United States
- Glycobiology Research and Training Center, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
24
|
Abassi Z, Armaly Z, Heyman SN. Glycocalyx Degradation in Ischemia-Reperfusion Injury. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:752-767. [PMID: 32035883 DOI: 10.1016/j.ajpath.2019.08.019] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/13/2019] [Accepted: 08/20/2019] [Indexed: 02/06/2023]
Abstract
The glycocalyx is a layer coating the luminal surface of vascular endothelial cells. It is vital for endothelial function as it participates in microvascular reactivity, endothelium interaction with blood constituents, and vascular permeability. Structural and functional damage to glycocalyx occurs in various disease states. A prominent clinical situation characterized by glycocalyx derangement is ischemia-reperfusion (I/R) of the whole body as well as during selective I/R to organs such as the kidney, heart, lung, or liver. Degradation of the glycocalyx is now considered a cornerstone in I/R-related endothelial dysfunction, which further impairs local microcirculation with a feed-forward loop of organ damage, due to vasoconstriction, leukocyte adherence, and activation of the immune response. Glycocalyx damage during I/R is evidenced by rising plasma levels of its principal constituents, heparan sulfate and syndecan-1. By contrast, the concentrations of these compounds in the circulation decrease after successful protective interventions in I/R, suggesting their use as surrogate biomarkers of endothelial integrity. In light of the importance of the glycocalyx in preserving endothelial cell integrity and its involvement in pathologic conditions, several promising therapeutic strategies to restore the damaged glycocalyx and to attenuate its deleterious consequences have been suggested. This review focuses on alterations of glycocalyx during I/R injury in general (to vital organs in particular), and on maneuvers aimed at glycocalyx recovery during I/R injury.
Collapse
Affiliation(s)
- Zaid Abassi
- Department of Physiology, The Ruth & Bruce Rappaport Faculty of Medicine, Technion-Israeli Institute of Technology, Haifa, Israel; Laboratory Medicine, Rambam Health Campus, Haifa, Israel.
| | - Zaher Armaly
- Department of Nephrology, Nazareth Hospital, Nazareth, Azrieli Faculty of Medicine-Bar Ilan University, Jerusalem, Israel
| | - Samuel N Heyman
- Department of Medicine, Hadassah Hebrew University Hospital, Mt. Scopus, Jerusalem, Israel
| |
Collapse
|
25
|
Abassi Z, Goligorsky MS. Heparanase in Acute Kidney Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:685-702. [PMID: 32274732 PMCID: PMC7369981 DOI: 10.1007/978-3-030-34521-1_28] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Recent years have brought about fledgling realization of the role played by heparanase in the pathogenesis of diverse diseases including kidney diseases and, specifically, acute kidney injury. Human heparanase-1 is critically and uniquely engaged in cleavage of heparan sulfate, an integral part of glycocalyx and extracellular matrix where it harbors distinct growth factors, cytokines, and other biologically active molecules. The enzyme is induced and activated in acute kidney injury regardless of its causes, ischemic, nephrotoxic, septic or transplantation-related. This event unleashes a host of sequelae characteristic of the pathogenesis of acute kidney injury, such as induction and reinforcement of innate immune responses, predisposition to thrombosis, activation of monocytes/macrophages and remodeling of the extracellular matrix, thus setting up the stage for future fibrotic complications and development of chronic kidney disease. We briefly discuss the emerging therapeutic strategies of inhibiting heparanase, as well as the diagnostic value of detecting products of heparanase activity for prognostication and treatment.
Collapse
Affiliation(s)
- Zaid Abassi
- Department of Physiology, Bruce Rappaport Faculty of Medicine, Technion-IIT, Haifa, Israel.
- Department of Laboratory Medicine, Rambam Health Care Campus, Haifa, Israel.
| | - M S Goligorsky
- Departments of Medicine, Physiology and Pharmacology, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
26
|
Role of Heparanase in Macrophage Activation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:445-460. [PMID: 32274721 DOI: 10.1007/978-3-030-34521-1_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Macrophages represent one of the most diverse immunocyte populations, constantly shifting between various phenotypes/functional states. In addition to execution of vital functions in normal physiological conditions, macrophages represent a key contributing factor in the pathogenesis of some of the most challenging diseases, such as chronic inflammatory disorders, diabetes and its complications, and cancer. Macrophage polarization studies focus primarily on cytokine-mediated mechanisms. However, to explore the full spectrum of macrophage action, additional, non-cytokine pathways responsible for altering macrophage phenotype have to be taken into consideration as well. Heparanase, the only known mammalian endoglycosidase that cleaves heparan sulfate glycosaminoglycans, has been shown to contribute to the altered macrophage phenotypes in vitro and in numerous animal models of inflammatory conditions, occurring either in the presence of microbial products or in the setting of non-infectious "aseptic" inflammation. Here we discuss the involvement of heparanase in shaping macrophage responses and provide information that may help to establish the rationale for heparanase-targeting interventions aimed at preventing abnormal macrophage activation in various disorders.
Collapse
|
27
|
van der Vlag J, Buijsers B. Heparanase in Kidney Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1221:647-667. [PMID: 32274730 DOI: 10.1007/978-3-030-34521-1_26] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The primary filtration of blood occurs in the glomerulus in the kidney. Destruction of any of the layers of the glomerular filtration barrier might result in proteinuric disease. The glomerular endothelial cells and especially its covering layer, the glycocalyx, play a pivotal role in development of albuminuria. One of the main sulfated glycosaminoglycans in the glomerular endothelial glycocalyx is heparan sulfate. The endoglycosidase heparanase degrades heparan sulfate, thereby affecting glomerular barrier function, immune reactivity and inflammation. Increased expression of glomerular heparanase correlates with loss of glomerular heparan sulfate in many glomerular diseases. Most importantly, heparanase knockout in mice prevented the development of albuminuria after induction of experimental diabetic nephropathy and experimental glomerulonephritis. Therefore, heparanase could serve as a pharmacological target for glomerular diseases. Several factors that regulate heparanase expression and activity have been identified and compounds aiming to inhibit heparanase activity are currently explored.
Collapse
Affiliation(s)
- Johan van der Vlag
- Department of Nephrology (480), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands.
| | - Baranca Buijsers
- Department of Nephrology (480), Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA, Nijmegen, The Netherlands
| |
Collapse
|
28
|
Goldberg R, Meirovitz A, Abecassis A, Hermano E, Rubinstein AM, Nahmias D, Grinshpun A, Peretz T, Elkin M. Regulation of Heparanase in Diabetes-Associated Pancreatic Carcinoma. Front Oncol 2019; 9:1405. [PMID: 31921662 PMCID: PMC6914686 DOI: 10.3389/fonc.2019.01405] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 11/27/2019] [Indexed: 12/18/2022] Open
Abstract
While at least six types of cancer have been associated with diabetes, pancreatic ductal adenocarcinoma (PDAC) and diabetes exhibit a unique bidirectional relationship. Recent reports indicate that majority of PDAC patients display hyperglycemia, and ~50% have concurrent diabetes. In turn, hyperglycemic/diabetic state in PDAC patients fosters enhanced growth and dissemination of the tumor. Heparanase enzyme (the sole mammalian endoglycosidase degrading glycosaminoglycan heparan sulfate) is tightly implicated in PDAC progression, aggressiveness, and therapy resistance. Overexpression of heparanase is a characteristic feature of PDAC, correlating with poor prognosis. However, given the lack of heparanase expression in normal pancreatic tissue, the regulatory mechanisms responsible for induction of the enzyme in PDAC have remained largely unknown. Previously reported inducibility of heparanase gene by diabetic milieu components in several non-cancerous cell types prompted us to hypothesize that in the setting of diabetes-associated PDAC, hyperglycemic state may induce heparanase overexpression. Here, utilizing a mouse model of diet-induced metabolic syndrome/diabetes, we found accelerated PDAC progression in hyperglycemic mice, occurring along with induction of heparanase in PDAC. In vitro, we demonstrated that advanced glycation end-products (AGE), which are largely thought as oxidative derivatives resulting from chronic hyperglycemia, and the receptor for AGE (RAGE) are responsible for heparanase induction in PDAC cells. These findings underscore the new mechanism underlying preferential expression of heparanase in pancreatic cancer. Moreover, taken together with the well-established causal role of the enzyme in PDAC progression, our findings indicate that heparanase may sustain (at least in part) reciprocal causality between diabetes and pancreatic tumorigenesis.
Collapse
Affiliation(s)
- Rachel Goldberg
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amichay Meirovitz
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Alexia Abecassis
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Esther Hermano
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ariel M Rubinstein
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Daniela Nahmias
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Albert Grinshpun
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Tamar Peretz
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Michael Elkin
- Sharett Oncology Institute, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
29
|
Abstract
The kidney harbours different types of endothelia, each with specific structural and functional characteristics. The glomerular endothelium, which is highly fenestrated and covered by a rich glycocalyx, participates in the sieving properties of the glomerular filtration barrier and in the maintenance of podocyte structure. The microvascular endothelium in peritubular capillaries, which is also fenestrated, transports reabsorbed components and participates in epithelial cell function. The endothelium of large and small vessels supports the renal vasculature. These renal endothelia are protected by regulators of thrombosis, inflammation and complement, but endothelial injury (for example, induced by toxins, antibodies, immune cells or inflammatory cytokines) or defects in factors that provide endothelial protection (for example, regulators of complement or angiogenesis) can lead to acute or chronic renal injury. Moreover, renal endothelial cells can transition towards a mesenchymal phenotype, favouring renal fibrosis and the development of chronic kidney disease. Thus, the renal endothelium is both a target and a driver of kidney and systemic cardiovascular complications. Emerging therapeutic strategies that target the renal endothelium may lead to improved outcomes for both rare and common renal diseases.
Collapse
|
30
|
Hermano E, Goldberg R, Rubinstein AM, Sonnenblick A, Maly B, Nahmias D, Li JP, Bakker MAH, van der Vlag J, Vlodavsky I, Peretz T, Elkin M. Heparanase Accelerates Obesity-Associated Breast Cancer Progression. Cancer Res 2019; 79:5342-5354. [PMID: 31481501 DOI: 10.1158/0008-5472.can-18-4058] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 06/06/2019] [Accepted: 08/26/2019] [Indexed: 11/16/2022]
Abstract
Obese women have higher risk of bearing breast tumors that are highly aggressive and resistant to therapies. Tumor-promoting effects of obesity occur locally via adipose inflammation and related alterations to the extracellular matrix (ECM) as well as systemically via circulating metabolic mediators (e.g., free fatty acids, FFA) associated with excess adiposity and implicated in toll-like receptor-mediated activation of macrophages-key cellular players in obesity-related cancer progression. Although the contribution of macrophages to proneoplastic effects of obesity is well documented, the role of ECM components and their enzymatic degradation is less appreciated. We show that heparanase, the sole mammalian endoglucuronidase that cleaves heparan sulfate in ECM, is preferentially expressed in clinical/experimental obesity-associated breast tumors. Heparanase deficiency abolished obesity-accelerated tumor progression in vivo. Heparanase orchestrated a complex molecular program that occurred concurrently in adipose and tumor tissue and sustained the cancer-promoting action of obesity. Heparanase was required for adipose tissue macrophages to produce inflammatory mediators responsible for local induction of aromatase, a rate-limiting enzyme in estrogen biosynthesis. Estrogen upregulated heparanase in hormone-responsive breast tumors. In subsequent stages, elevated levels of heparanase induced acquisition of procancerous phenotype by tumor-associated macrophages, resulting in activation of tumor-promoting signaling and acceleration of breast tumor growth under obese conditions. As techniques to screen for heparanase expression in tumors become available, these findings provide rational and a mechanistic basis for designing antiheparanase approaches to uncouple obesity and breast cancer in a rapidly growing population of obese patients. SIGNIFICANCE: This study reveals the role of heparanase in promoting obesity-associated breast cancer and provides a mechanistically informed approach to uncouple obesity and breast cancer in a rapidly growing population of obese patients.
Collapse
Affiliation(s)
- Esther Hermano
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Rachel Goldberg
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ariel M Rubinstein
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Amir Sonnenblick
- Oncology Division, Tel Aviv Sourasky Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Bella Maly
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Daniela Nahmias
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Jin-Ping Li
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Marinka A H Bakker
- Nephrology Research Laboratory, Department of Nephrology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Johan van der Vlag
- Nephrology Research Laboratory, Department of Nephrology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands
| | - Israel Vlodavsky
- Cancer and Vascular Biology Research Center, The Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Tamar Peretz
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
- Hebrew University Medical School, Jerusalem, Israel
| | - Michael Elkin
- Sharett Institute of Oncology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.
- Hebrew University Medical School, Jerusalem, Israel
| |
Collapse
|
31
|
Masola V, Zaza G, Gambaro G, Franchi M, Onisto M. Role of heparanase in tumor progression: Molecular aspects and therapeutic options. Semin Cancer Biol 2019; 62:86-98. [PMID: 31348993 DOI: 10.1016/j.semcancer.2019.07.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 07/11/2019] [Accepted: 07/22/2019] [Indexed: 02/08/2023]
Abstract
Heparanase (HPSE) is an endoglycosidase that catalyses the cutting of the side chains of heparan-sulphate proteoglycans (HS), thus determining the remodelling of the extracellular matrix and basement membranes, as well as promoting the release of different HS-related molecules as growth factors, cytokines and enzymes. Ever since the HPSE was identified in the late 1980s, several experimental studies have shown that its overexpression was instrumental in increasing tumor growth, metastatic dissemination, angiogenesis and inflammation. More recently, HPSE involvment has also been demonstrated in mediating tumor-host crosstalk, in inducing gene transcription, in the activation of signaling pathways and in the formation of exosomes and in autophagy. All of these activities (enzymatic and non-enzymatic) together make heparanase a multifunctional molecule that increases the aggressiveness and chemo-resistance of tumor cells. Conversely, heparanase gene-silencing or tumor treatment with compounds that inhibit heparanase activity have been shown to significantly attenuate tumor progression in different animal models of tumorigenesis, further emphasizing the therapeutic potential of anti-heparanase therapy for several types of neoplasms. This review focuses on present knowledge and recent development in the study of heparanase in cancer progression as well as on novel mechanisms by which heparanase regulates tumor metastasis and chemo-resistance. Moreover, recent advances in strategies for its inhibition as a potential therapeutic option will be discussed.
Collapse
Affiliation(s)
- Valentina Masola
- Dept. of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy; Dept. of Medicine, University of Verona, 37134, Verona, Italy
| | - Gianluigi Zaza
- Dept. of Medicine, University of Verona, 37134, Verona, Italy
| | | | - Marco Franchi
- Dept. of Life Quality Sciences, University of Bologna, Corso D'Augusto 237, 47921, Rimini, Italy
| | - Maurizio Onisto
- Dept. of Biomedical Sciences, University of Padova, Viale G. Colombo 3, 35121, Padova, Italy.
| |
Collapse
|
32
|
Li J, Qiu P, Wang S, Wu J, He Q, Li K, Xu L. β-N-Oxalyl-L-α,β-diaminopropionic acid from Panax notoginseng plays a major role in the treatment of type 2 diabetic nephropathy. Biomed Pharmacother 2019; 114:108801. [PMID: 30928803 DOI: 10.1016/j.biopha.2019.108801] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/18/2019] [Accepted: 03/18/2019] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is one of the most serious and dangerous chronic complications of diabetes mellitus.Panax notoginseng has been widely used with great efficacy in the long-term treatment of kidney disease. However, the mechanism by which it exerts its effects has not been fully elucidated. AIM We sought to identify the major components ofPanax notoginseng that are effective in reducing the symptoms of DN in vitro and in vivo. METHODS Inhibition of cell proliferation and collagen secretion were used to screen the ten most highly concentrated components ofPanax notoginseng. The STZ-induced DN rat model on a high-fat-high-glucose diet was used to investigate the renal protective effect of Panax notoginseng and dencichine and their underlying molecular mechanisms. RESULTS Among the ten components analysed, dencichine (β-N-oxalyl-L-α,β-diaminopropionic acid) was the most protective against DN. Dencichine andPanax notoginseng attenuated glucose and lipid metabolic disorders in STZ-induced DN rats on a high-fat-high-glucose diet. In the untreated DN rats, we observed albuminuria, renal failure, and pathological changes. However, treatment with dencichine and Panax notoginseng alleviated these symptoms. We also observed that dencichine suppressed the expression of TGF-β1 and Smad2/3, which mediates mesangial cell proliferation and extracellular matrix (ECM) accumulation in the glomerulus, and enhanced the expression of Smad7, the endogenous inhibitor of the TGF-β1/Smad signalling pathway. CONCLUSION From these results, we concluded that dencichine is the main compound inPanax notoginseng that is responsible for alleviating renal injury in the experimental DN model. Its mechanism may be related to the reduction of the deposition of ECM in glomeruli and inhibition of the epithelial mesenchymal transformation (EMT) by inhibition of the TGF-β1/Smad signalling pathway.
Collapse
Affiliation(s)
- Jie Li
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, 710032, Xi'an, China
| | - Pengcheng Qiu
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, 710032, Xi'an, China
| | - Siwang Wang
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, 710032, Xi'an, China; School of Life Sciences, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China.
| | - Junsheng Wu
- School of Life Sciences, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Qiaoyan He
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, 710032, Xi'an, China
| | - Kaifeng Li
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, 710032, Xi'an, China
| | - Lu Xu
- Department of Chinese Materia Medica and Natural Medicines, School of Pharmacy, Air Force Medical University, 710032, Xi'an, China
| |
Collapse
|
33
|
Zhang X, Sun D, Song JW, Zullo J, Lipphardt M, Coneh-Gould L, Goligorsky MS. Endothelial cell dysfunction and glycocalyx – A vicious circle. Matrix Biol 2018; 71-72:421-431. [DOI: 10.1016/j.matbio.2018.01.026] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 12/16/2022]
|
34
|
An X, Zhang M, Zhou S, Lu T, Chen Y, Yao L. Xiao-Shen-Formula, a Traditional Chinese Medicine, Improves Glomerular Hyper-Filtration in Diabetic Nephropathy via Inhibiting Arginase Activation and Heparanase Expression. Front Physiol 2018; 9:1195. [PMID: 30319431 PMCID: PMC6169603 DOI: 10.3389/fphys.2018.01195] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 08/08/2018] [Indexed: 11/13/2022] Open
Abstract
Hyperglycemia induces glomerular hyper-filtration, which contributes to the development of diabetic nephropathy (DN), a condition that remains a challenge for treatment. The present study investigated the effect of Xiao-Shen-Formula (XSF) used for treatment of renal injury in type 1 DN mice model induced by streptozotocin (STZ) and its underlying mechanism in cultured human glomerular endothelial cell (hGECs). Studies were performed using control, diabetic DN, DN treated with XSF groups (1 g/kg/d, LXSF or 3 g/kg/d, HXSF) for 6 weeks and hGECs were post-treated with mice serum containing HXSF (MS-HXSF) and arginase inhibitor (ABH, 100 μM) in high glucose medium. HXSF treatment restored STZ-induced renal hyper-filtration, glomerulosclerosis, renal microvascular remodeling and the increased levels of systemic reactive oxidative species and inflammatory cytokines, accompanied by preventing the decreased expression of glomerular heparin sulfate and the increased levels of cortical heparanase and argianse2 protein and arginase activity. In hGECs study, MS-HXSF ameliorated the enhancement in arginase activity, the protein/mRNA expression of heparanase, mRNA levels of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, monocyte chemoattractant protein-1 and permeability of hGECs monolayers as well as the depression of nitric oxide production. Besides all these protective effects, XSF blunted the mRNA expression of TNF-α in vivo and vitro studies as well, which was not changed by the post-treatment of ABH or HXSF plus ABH. This study demonstrated that the protective effect of XSF might be related with vascular prevention, anti-inflammation and anti-oxidation through intervening multi-targets including glomerular endothelial arginase-heparanase signaling pathway in DN model.
Collapse
Affiliation(s)
- Xiaofei An
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Maoxiang Zhang
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China.,Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sisi Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tian Lu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongjun Chen
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China.,Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Yao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China.,South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
35
|
Zhao Y, Liu J, Ten S, Zhang J, Yuan Y, Yu J, An X. Plasma heparanase is associated with blood glucose levels but not urinary microalbumin excretion in type 2 diabetic nephropathy at the early stage. Ren Fail 2018; 39:698-701. [PMID: 28994624 PMCID: PMC6446149 DOI: 10.1080/0886022x.2017.1384391] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Aim: To explore the possible correlations between plasma heparanase and albuminuria, glucose and lipid metabolism in the type 2 diabetic nephropathy patients at the early stage. Methods: One hundred and forty patients with type 2 diabetic nephropathy at early stage were recruited into the study. Plasma heparanase and the characterized advanced glycation end products (AGEs), carboxymethyllysine (CML) were measured by enzyme-linked immunosorbent assay. Results: Plasma heparanase was positively associated with fasting blood glucose (R = 0.24, p = .01) while heparanase was not significantly correlated with the urinary microalbumin to creatinine ratio (urinary mAlb/Cr) (R = 0.05, p = .58) and CML (R = 0.16, p = .26). On stepwise linear regression analysis, fasting blood glucose was the main independent determinants of plasma heparanase concentration. Conclusion: Plasma heparanase is not significantly associated with urinary mAlb/Cr while it is correlated positively with blood glucose levels in the early stage of diabetic nephropathy. Plasma heparnase might be regarded as a marker for vascular endothelial cells injury in diabetic patients.
Collapse
Affiliation(s)
- Yue Zhao
- a Department of Endocrinology , Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China
| | - Jingshun Liu
- a Department of Endocrinology , Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China
| | - Shichao Ten
- a Department of Endocrinology , Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China
| | - Jisheng Zhang
- b Department of Otorhinolaryngology , Affiliated Hospital of Qingdao University , Qingdao , Shandong , China
| | - Yanggang Yuan
- c Department of Nephrology , The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province People's Hospital , Nanjing , Jiangsu , China
| | - Jiangyi Yu
- a Department of Endocrinology , Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China
| | - Xiaofei An
- a Department of Endocrinology , Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China
| |
Collapse
|
36
|
Muhammad RS, Abu-Saleh N, Kinaneh S, Agbaria M, Sabo E, Grajeda-Iglesias C, Volkova N, Hamoud S. Heparanase inhibition attenuates atherosclerosis progression and liver steatosis in E 0 mice. Atherosclerosis 2018; 276:155-162. [PMID: 30075439 DOI: 10.1016/j.atherosclerosis.2018.07.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 07/01/2018] [Accepted: 07/18/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND AND AIMS Increased oxidative stress is associated with accelerated atherosclerosis. Emerging evidence highlights the role of heparanase in atherogenesis, where heparanase inhibitor PG545 reduces oxidative stress in apolipoprotein E deficient mice (E0 mice). Herein, we studied the effects of PG545 on atherosclerosis progression in E0 mice. METHODS Male E0 mice fed a high-fat diet (n = 20) were divided into 3 groups treated with weekly intraperitoneal injections of either low (0.2 mg/mouse) or high dose (0.4 mg/mouse)PG545 or normal saline (controls) for twelve weeks. Body weight and food intake were measured weekly. At the end of the treatment period, blood pressure was measured, animals were sacrificed and serum samples were collected and assessed for biochemical parameters and oxidative stress. Aortic vessels and livers were collected for atherosclerotic plaques and histopathological analysis, respectively. RESULTS Blood pressure decreased in mice treated with low, but not high dose of PG545. In addition, heparanase inhibition caused a dose-dependent reduction in serum oxidative stress, total cholesterol, low-density lipoproteins, triglycerides, high-density lipoproteins, and aryl esterase activity. Although food intake was not reduced by PG545, body weight gain was significantly attenuated in PG545 treated groups. Both doses of PG545 caused a marked reduction in aortic wall thickness and atherosclerosis development, and liver steatosis. Liver enzymes and serum creatinine were not affected by PG545. CONCLUSIONS Heparanase inhibition by PG545 caused a significant reduction in lipid profile and serum oxidative stress along with attenuation of atherosclerosis, aortic wall thickness, and liver steatosis. Moreover, PG545 attenuated weight gain without reducing food intake. Collectively, these findings suggest that heparanase blockade is highly effective in slowing atherosclerosis formation and progression, and decreasing liver steatosis.
Collapse
Affiliation(s)
- Rabia Shekh Muhammad
- Department of Internal Medicine E, Rambam Health Care Campus and Rappaport Faculty of Medicine Haifa, Israel
| | - Niroz Abu-Saleh
- Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Safa Kinaneh
- Department of Physiology, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Mohammad Agbaria
- Department of Internal Medicine A, Rambam Health Care Campus, Haifa, Israel
| | - Edmond Sabo
- Department of Pathology, Rambam Health Care Campus and Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | | | - Nina Volkova
- Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Shadi Hamoud
- Department of Internal Medicine E, Rambam Health Care Campus and Rappaport Faculty of Medicine Haifa, Israel; Lipid Research Laboratory, Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| |
Collapse
|
37
|
An X, Zhang L, Yao Q, Li L, Wang B, Zhang J, He M, Zhang J. The receptor for advanced glycation endproducts mediates podocyte heparanase expression through NF-κB signaling pathway. Mol Cell Endocrinol 2018; 470:14-25. [PMID: 28478303 DOI: 10.1016/j.mce.2017.05.004] [Citation(s) in RCA: 12] [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] [Received: 02/26/2017] [Revised: 05/02/2017] [Accepted: 05/02/2017] [Indexed: 01/18/2023]
Abstract
Heparanase degrades heparan sulfate in glomerular basement membrane (GBM) and plays an important role in diabetic nephropathy (DN). However, its regulating mechanisms remain to be deciphered. Our present study showed that the major advanced glycation endproducts (AGEs), CML-BSA, significantly increased heparanase expression in cultured podocytes and the effect was blocked by the receptor for advanced glycation endproducts (RAGE) knockdown, antibody and antagonist. In addition, NF-κB p65 phosphorylation was elevated and the increased heparanase expression and secretion upon CML-BSA could be attenuated by NF-κB inhibitor PDTC. Mechanistically, CML-BSA activated heparanase promoter through p65 directly binding to its promoter. Furthermore, the in vivo study showed that serum and renal cortex AGEs levels, glomerular p65 phosphorylation and heparanase expression were significantly increased in DN mice. Taken together, our data suggest that AGEs and RAGE interaction increases podocyte heparanase expression by activating NF-κB signal pathway, which is involved in GBM damages of DN.
Collapse
Affiliation(s)
- Xiaofei An
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Lin Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Qiuming Yao
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Ling Li
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Bin Wang
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China
| | - Jisheng Zhang
- Department of Otorhinolaryngology, Affiliated Hospital of Qingdao University, Qingdao 266003, China.
| | - Ming He
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China.
| | - Jinan Zhang
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai 201508, China.
| |
Collapse
|
38
|
Abassi Z, Hamoud S, Hassan A, Khamaysi I, Nativ O, Heyman SN, Muhammad RS, Ilan N, Singh P, Hammond E, Zaza G, Lupo A, Onisto M, Bellin G, Masola V, Vlodavsky I, Gambaro G. Involvement of heparanase in the pathogenesis of acute kidney injury: nephroprotective effect of PG545. Oncotarget 2018; 8:34191-34204. [PMID: 28388547 PMCID: PMC5470960 DOI: 10.18632/oncotarget.16573] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 03/16/2017] [Indexed: 11/29/2022] Open
Abstract
Despite the high prevalence of acute kidney injury (AKI) and its association with increased morbidity and mortality, therapeutic approaches for AKI are disappointing. This is largely attributed to poor understanding of the pathogenesis of AKI. Heparanase, an endoglycosidase that cleaves heparan sulfate, is involved in extracellular matrix turnover, inflammation, kidney dysfunction, diabetes, fibrosis, angiogenesis and cancer progression. The current study examined the involvement of heparanase in the pathogenesis of ischemic reperfusion (I/R) AKI in a mouse model and the protective effect of PG545, a potent heparanase inhibitor. I/R induced tubular damage and elevation in serum creatinine and blood urea nitrogen to a higher extent in heparanase over-expressing transgenic mice vs. wild type mice. Moreover, TGF-β, vimentin, fibronectin and α-smooth muscle actin, biomarkers of fibrosis, and TNFα, IL6 and endothelin-1, biomarkers of inflammation, were upregulated in I/R induced AKI, primarily in heparanase transgenic mice, suggesting an adverse role of heparanase in the pathogenesis of AKI. Remarkably, pretreatment of mice with PG545 abolished kidney dysfunction and the up-regulation of heparanase, pro-inflammatory (i.e., IL-6) and pro-fibrotic (i.e., TGF-β) genes induced by I/R. The present study provides new insights into the involvement of heparanase in the pathogenesis of ischemic AKI. Our results demonstrate that heparanase plays a deleterious role in the development of renal injury and kidney dysfunction, attesting heparanase inhibition as a promising therapeutic approach for AKI.
Collapse
Affiliation(s)
- Zaid Abassi
- Department of Physiology, The Rappaport Faculty of Medicine, Technion, Haifa, Israel.,Department of Laboratory Medicine, Rambam Health Care Campus, Haifa, Israel
| | - Shadi Hamoud
- Department of Internal Medicine E, Rambam Health Care Campus, Haifa, Israel
| | - Ahmad Hassan
- Department of Internal Medicine A, Rambam Health Care Campus, Haifa, Israel
| | - Iyad Khamaysi
- Department of Gastroenterology, Rambam Health Care Campus, Haifa, Israel
| | - Omri Nativ
- Department of Physiology, The Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Samuel N Heyman
- Department of Internal Medicine, Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | | | - Neta Ilan
- Department of Cancer and Vascular Biology Research Center, The Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Preeti Singh
- Department of Cancer and Vascular Biology Research Center, The Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | | | | | - Antonio Lupo
- Department of Medicine, Renal Unit, Verona, Italy
| | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Catholic University of the Sacred Heart, Roma, Italy
| | | | | | - Israel Vlodavsky
- Department of Cancer and Vascular Biology Research Center, The Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Giovani Gambaro
- Department of Medicine, Columbus-Gemelli Hospital, Catholic University of the Sacred Heart, Roma, Italy
| |
Collapse
|
39
|
Simeonovic CJ, Popp SK, Starrs LM, Brown DJ, Ziolkowski AF, Ludwig B, Bornstein SR, Wilson JD, Pugliese A, Kay TWH, Thomas HE, Loudovaris T, Choong FJ, Freeman C, Parish CR. Loss of intra-islet heparan sulfate is a highly sensitive marker of type 1 diabetes progression in humans. PLoS One 2018; 13:e0191360. [PMID: 29415062 PMCID: PMC5802856 DOI: 10.1371/journal.pone.0191360] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/03/2018] [Indexed: 12/20/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which insulin-producing beta cells in pancreatic islets are progressively destroyed. Clinical trials of immunotherapies in recently diagnosed T1D patients have only transiently and partially impacted the disease course, suggesting that other approaches are required. Our previous studies have demonstrated that heparan sulfate (HS), a glycosaminoglycan conventionally expressed in extracellular matrix, is present at high levels inside normal mouse beta cells. Intracellular HS was shown to be critical for beta cell survival and protection from oxidative damage. T1D development in Non-Obese Diabetic (NOD) mice correlated with loss of islet HS and was prevented by inhibiting HS degradation by the endoglycosidase, heparanase. In this study we investigated the distribution of HS and heparan sulfate proteoglycan (HSPG) core proteins in normal human islets, a role for HS in human beta cell viability and the clinical relevance of intra-islet HS and HSPG levels, compared to insulin, in human T1D. In normal human islets, HS (identified by 10E4 mAb) co-localized with insulin but not glucagon and correlated with the HSPG core proteins for collagen type XVIII (Col18) and syndecan-1 (Sdc1). Insulin-positive islets of T1D pancreases showed significant loss of HS, Col18 and Sdc1 and heparanase was strongly expressed by islet-infiltrating leukocytes. Human beta cells cultured with HS mimetics showed significantly improved survival and protection against hydrogen peroxide-induced death, suggesting that loss of HS could contribute to beta cell death in T1D. We conclude that HS depletion in beta cells, possibly due to heparanase produced by insulitis leukocytes, may function as an important mechanism in the pathogenesis of human T1D. Our findings raise the possibility that intervention therapy with dual activity HS replacers/heparanase inhibitors could help to protect the residual beta cell mass in patients recently diagnosed with T1D.
Collapse
Affiliation(s)
- Charmaine J. Simeonovic
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
- * E-mail:
| | - Sarah K. Popp
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Lora M. Starrs
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Debra J. Brown
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Andrew F. Ziolkowski
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Barbara Ludwig
- Department of Internal Medicine III, Carl Gustav Carus Medical School, Technical University of Dresden, Dresden, Germany
| | - Stefan R. Bornstein
- Department of Internal Medicine III, Carl Gustav Carus Medical School, Technical University of Dresden, Dresden, Germany
| | - J. Dennis Wilson
- Department of Endocrinology, The Canberra Hospital, Woden, Australian Capital Territory, Australia
| | - Alberto Pugliese
- Diabetes Research Institute, Departments of Medicine, Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Thomas W. H. Kay
- St Vincent’s Institute of Medical Research, Fitzroy, Melbourne, Victoria, Australia
| | - Helen E. Thomas
- St Vincent’s Institute of Medical Research, Fitzroy, Melbourne, Victoria, Australia
| | - Thomas Loudovaris
- St Vincent’s Institute of Medical Research, Fitzroy, Melbourne, Victoria, Australia
| | - Fui Jiun Choong
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Craig Freeman
- Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| | - Christopher R. Parish
- Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
40
|
Sell DR, Nemet I, Liang Z, Monnier VM. Evidence of glucuronidation of the glycation product LW-1: tentative structure and implications for the long-term complications of diabetes. Glycoconj J 2018; 35:177-190. [PMID: 29305779 DOI: 10.1007/s10719-017-9810-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 12/01/2017] [Indexed: 01/06/2023]
Abstract
LW-1 is a collagen-linked blue fluorophore whose skin levels increase with age, diabetes and end-stage renal disease (ESRD), and correlate with the long-term progression of microvascular disease and indices of subclinical cardiovascular disease in type 1 diabetes. The chemical structure of LW-1 is still elusive, but earlier NMR analyses showed it has a lysine residue in an aromatic ring coupled to a sugar molecule reminiscent of advanced glycation end-products (AGEs). We hypothesized and demonstrate here that the unknown sugar is a N-linked glucuronic acid. LW-1 was extracted and highly purified from ~99 g insoluble skin collagen obtained at autopsy from patients with diabetes/ESRD using multiple rounds of proteolytic digestion and purification by liquid chromatography (LC). Advanced NMR techniques (1H-NMR, 13C-NMR, 1H-13C HSQC, 1H-1H TOCSY, 1H-13C HMBC) together with LC-mass spectrometry (MS) revealed a loss of 176 amu (atomic mass unit) unequivocally point to the presence of a glucuronic acid moiety in LW-1. To confirm this data, LW-1 was incubated with β-glycosidases (glucosidase, galactosidase, glucuronidase) and products were analyzed by LC-MS. Only glucuronidase could cleave the sugar from the parent molecule. These results establish LW-1 as a glucuronide, now named glucuronidine, and for the first time raise the possible existence of a "glucuronidation pathway of diabetic complications". Future research is needed to rigorously probe this concept and elucidate the molecular origin and biological source of a circulating glucuronidine aglycone.
Collapse
Affiliation(s)
- David R Sell
- Department of Pathology, Case Western Reserve University, Wolstein Research Bldg. 5-301, 2103 Cornell Road, Cleveland, OH, 44106, USA.
| | - Ina Nemet
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, 44195, USA
| | - Zhili Liang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Vincent M Monnier
- Department of Pathology, Case Western Reserve University, Wolstein Research Bldg. 5-301, 2103 Cornell Road, Cleveland, OH, 44106, USA. .,Department of Biochemistry, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
41
|
Masola V, Zaza G, Bellin G, Dall'Olmo L, Granata S, Vischini G, Secchi MF, Lupo A, Gambaro G, Onisto M. Heparanase regulates the M1 polarization of renal macrophages and their crosstalk with renal epithelial tubular cells after ischemia/reperfusion injury. FASEB J 2018; 32:742-756. [PMID: 28970256 DOI: 10.1096/fj.201700597r] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Heparanase (HPSE) is part of the biologic network triggered by ischemia/reperfusion (I/R) injury, a complication of renal transplantation and acute kidney injury. During this period, the kidney or graft undergoes a process of macrophages recruitment and activation. HPSE may therefore control these biologic effects. We measured the ability of HPSE and its inhibitor, SST0001, to regulate macrophage polarization and the crosstalk between macrophages and HK-2 renal tubular cells during in vitro hypoxia/reoxygenation (H/R). Furthermore, we evaluated in vivo renal inflammation, macrophage polarization, and histologic changes in mice subjected to monolateral I/R and treated with SST0001 for 2 or 7 d. The in vitro experiments showed that HPSE sustained M1 macrophage polarization and modulated apoptosis, the release of damage associated molecular patterns in post-H/R tubular cells, the synthesis of proinflammatory cytokines, and the up-regulation of TLRs on both epithelial cells and macrophages. HPSE also regulated M1 polarization induced by H/R-injured tubular cells and the partial epithelial-mesenchymal transition of these epithelial cells by M1 macrophages. All these effects were prevented by inhibiting HPSE. Furthermore, the inhibition of HPSE in vivo reduced inflammation and M1 polarization in mice undergoing I/R injury, partially restored renal function and normal histology, and reduced apoptosis. These results show for the first time that HPSE regulates macrophage polarization as well as renal damage and repair after I/R. HPSE inhibitors could therefore provide a new pharmacologic approach to minimize acute kidney injury and to prevent the chronic profibrotic damages induced by I/R.-Masola, V., Zaza, G., Bellin, G., Dall'Olmo, L., Granata, S., Vischini, G., Secchi, M. F., Lupo, A., Gambaro, G., Onisto, M. Heparanase regulates the M1 polarization of renal macrophages and their crosstalk with renal epithelial tubular cells after ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Valentina Masola
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
| | - Gloria Bellin
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
| | - Luigi Dall'Olmo
- Azienda Ulss 3 Serenissima, Ospedale San Giovanni e Paolo, Venice, Italy
| | - Simona Granata
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
| | - Gisella Vischini
- Renal Unit, Università Cattolica del Sacro Cuore, Rome, Italy; and
| | | | - Antonio Lupo
- Renal Unit, Department of Medicine, University Hospital of Verona, Verona, Italy
| | - Giovanni Gambaro
- Renal Unit, Università Cattolica del Sacro Cuore, Rome, Italy; and
| | - Maurizio Onisto
- Department of Biomedical Sciences Padova, University of Padova, Padua, Italy
| |
Collapse
|
42
|
Farrugia BL, Lord MS, Melrose J, Whitelock JM. The Role of Heparan Sulfate in Inflammation, and the Development of Biomimetics as Anti-Inflammatory Strategies. J Histochem Cytochem 2018; 66:321-336. [PMID: 29290153 DOI: 10.1369/0022155417740881] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Key events that occur during inflammation include the recruitment, adhesion, and transmigration of leukocytes from the circulation to the site of inflammation. These events are modulated by chemokines, integrins, and selectins and the interaction of these molecules with glycosaminoglycans, predominantly heparan sulfate (HS). The development of HS/heparin mimetics that interfere or inhibit the interactions that occur between glycosaminoglycans and modulators of inflammation holds great potential for use as anti-inflammatory therapeutics. This review will detail the role of HS in the events that occur during inflammation, their interaction and modulation of inflammatory mediators, and the current advances in the development of HS/heparin mimetics as anti-inflammatory biotherapeutics.
Collapse
Affiliation(s)
- Brooke L Farrugia
- Graduate School of Biomedical Engineering, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - Megan S Lord
- Graduate School of Biomedical Engineering, University of New South Wales Sydney, Sydney, New South Wales, Australia
| | - James Melrose
- Graduate School of Biomedical Engineering, University of New South Wales Sydney, Sydney, New South Wales, Australia.,Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, New South Wales, Australia.,Sydney Medical School-Northern, Royal North Shore Hospital, The University of Sydney, St. Leonards, New South Wales, Australia
| | - John M Whitelock
- Graduate School of Biomedical Engineering, University of New South Wales Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
43
|
Secchi MF, Crescenzi M, Masola V, Russo FP, Floreani A, Onisto M. Heparanase and macrophage interplay in the onset of liver fibrosis. Sci Rep 2017; 7:14956. [PMID: 29097791 PMCID: PMC5668295 DOI: 10.1038/s41598-017-14946-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 10/18/2017] [Indexed: 12/26/2022] Open
Abstract
The heparan sulfate endoglycosidase heparanase (HPSE) is involved in tumor growth, chronic inflammation and fibrosis. Since a role for HPSE in chronic liver disease has not been demonstrated to date, the current study was aimed at investigating the involvement of HPSE in the pathogenesis of chronic liver injury. Herein, we revealed that HPSE expression increased in mouse livers after carbon tetrachloride (CCl4)-mediated chronic induction of fibrosis, but with a trend to decline during progression of the disease. In mouse fibrotic liver tissues HPSE immunostaining was restricted in necro-inflammatory areas, co-localizing with F4/80 macrophage marker and TNF-α. TNF-α treatment induced HPSE expression as well as HPSE secretion in U937 macrophages. Moreover, macrophage-secreted HPSE regulated the expression of α-SMA and fibronectin in hepatic stellate LX-2 cells. Finally, HPSE activity increased in the plasma of patients with liver fibrosis but it inversely correlated with liver stiffness. Our results suggest the involvement of HPSE in early phases of reaction to liver damage and inflammatory macrophages as an important source of HPSE. HPSE seems to play a key role in the macrophage-mediated activation of hepatic stellate cells (HSCs), thus suggesting that HPSE targeting could be a new therapeutic option in the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Maria Francesca Secchi
- University of Padova, Dept. Biomedical Sciences, 35121, Padova, Italy
- University of Padova, Dept. of Surgery, Oncology and Gastroenterology, 35124, Padova, Italy
| | - Marika Crescenzi
- University of Padova, Dept. of Surgery, Oncology and Gastroenterology, 35124, Padova, Italy
| | - Valentina Masola
- University of Padova, Dept. Biomedical Sciences, 35121, Padova, Italy
- University of Verona, Dept. of Medicine, 37134, Verona, Italy
| | - Francesco Paolo Russo
- University of Padova, Dept. of Surgery, Oncology and Gastroenterology, 35124, Padova, Italy.
| | - Annarosa Floreani
- University of Padova, Dept. of Surgery, Oncology and Gastroenterology, 35124, Padova, Italy
| | - Maurizio Onisto
- University of Padova, Dept. Biomedical Sciences, 35121, Padova, Italy.
| |
Collapse
|
44
|
Heparanase Inhibition Reduces Glucose Levels, Blood Pressure, and Oxidative Stress in Apolipoprotein E Knockout Mice. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7357495. [PMID: 29226146 PMCID: PMC5684525 DOI: 10.1155/2017/7357495] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 08/07/2017] [Accepted: 09/17/2017] [Indexed: 02/07/2023]
Abstract
Background Atherosclerosis is a multifactorial process. Emerging evidence highlights a role of the enzyme heparanase in various disease states, including atherosclerosis formation and progression. Objective The aim of the study was to investigate the effect of heparanase inhibition on blood pressure, blood glucose levels, and oxidative stress in apoE−/− mice. Methods Male apoE−/− mice were divided into two groups: one treated by the heparanase inhibitor PG545, administered intraperitoneally weekly for seven weeks, and the other serving as control group (injected with saline). Blood pressure was measured a day before sacrificing the animals. Serum glucose levels and lipid profile were measured. Assessment of oxidative stress was performed as well. Results PG545 significantly lowered blood pressure and serum glucose levels in treated mice. It also caused significant reduction of the serum oxidative stress. For safety concerns, liver enzymes were assessed, and PG545 caused significant elevation only of alanine aminotransferase, but not of the other hepatic enzymes. Conclusion Heparanase inhibition by PG545 caused marked reduction of blood pressure, serum glucose levels, and oxidative stress in apolipoprotein E deficient mice, possibly via direct favorable metabolic and hemodynamic changes caused by the inhibitor. Possible hepatotoxic and weight wasting effects are subject for future investigation.
Collapse
|
45
|
Boels MGS, Koudijs A, Avramut MC, Sol WMPJ, Wang G, van Oeveren-Rietdijk AM, van Zonneveld AJ, de Boer HC, van der Vlag J, van Kooten C, Eulberg D, van den Berg BM, IJpelaar DHT, Rabelink TJ. Systemic Monocyte Chemotactic Protein-1 Inhibition Modifies Renal Macrophages and Restores Glomerular Endothelial Glycocalyx and Barrier Function in Diabetic Nephropathy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2430-2440. [PMID: 28837800 DOI: 10.1016/j.ajpath.2017.07.020] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 06/26/2017] [Accepted: 07/05/2017] [Indexed: 11/25/2022]
Abstract
Inhibition of monocyte chemotactic protein-1 (MCP-1) with the Spiegelmer emapticap pegol (NOX-E36) shows long-lasting albuminuria-reducing effects in diabetic nephropathy. MCP-1 regulates inflammatory cell recruitment and differentiation of macrophages. Because the endothelial glycocalyx is also reduced in diabetic nephropathy, we hypothesized that MCP-1 inhibition restores glomerular barrier function through influencing macrophage cathepsin L secretion, thus reducing activation of the glycocalyx-degrading enzyme heparanase. Four weeks of treatment of diabetic Apoe knockout mice with the mouse-specific NOX-E36 attenuated albuminuria without any change in systemic hemodynamics, despite persistent loss of podocyte function. MCP-1 inhibition, however, increased glomerular endothelial glycocalyx coverage, with preservation of heparan sulfate. Mechanistically, both glomerular cathepsin L and heparanase expression were reduced. MCP-1 inhibition resulted in reduced CCR2-expressing Ly6Chi monocytes in the peripheral blood, without affecting overall number of kidney macrophages at the tissue level. However, the CD206+/Mac3+ cell ratio, as an index of presence of anti-inflammatory macrophages, increased in diabetic mice after treatment. Functional analysis of isolated renal macrophages showed increased release of IL-10, whereas tumor necrosis factor and cathepsin L release was reduced, further confirming polarization of tissue macrophages toward an anti-inflammatory phenotype during mouse-specific NOX-E36 treatment. We show that MCP-1 inhibition restores glomerular endothelial glycocalyx and barrier function and reduces tissue inflammation in the presence of ongoing diabetic injury, suggesting a therapeutic potential for NOX-E36 in diabetic nephropathy.
Collapse
Affiliation(s)
- Margien G S Boels
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Angela Koudijs
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - M Cristina Avramut
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Wendy M P J Sol
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Gangqi Wang
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Annemarie M van Oeveren-Rietdijk
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hetty C de Boer
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Cees van Kooten
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Bernard M van den Berg
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Daphne H T IJpelaar
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ton J Rabelink
- Einthoven Laboratory for Vascular and Regenerative Medicine, Department of Internal Medicine, Division of Nephrology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
46
|
Qin Q, Xu G, Qi W, Guo M, Wang Z, Xu W, Qiao Z, Gu Y, Niu J. Evaluation of the C-domain of heparanase during AGE-induced macrophage inflammatory response. Exp Ther Med 2017; 14:1017-1022. [PMID: 28810553 PMCID: PMC5526102 DOI: 10.3892/etm.2017.4609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 04/18/2017] [Indexed: 11/05/2022] Open
Abstract
Diabetic vasculopathy is intensified by macrophage inflammation caused by advanced glycation end products (AGEs). Heparanase (HPA) is a unique endoglycosidase, which cleaves heparan sulfate proteoglycans (HSPGs) including syndecan-1 (Syn-1) to further stimulate macrophage cell migration and inflammation. The present study was planned to evaluated the role of C-domain (if any) of HPA in AGE inflammatory response in macrophages. Cell viability was assessed using MTT assay, migration assay, ELISA for tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β) levels, mRNA expression by RT-PCR and heparan degrading enzyme assay for HPA activity. In the present study, we found that pretreatment with anti-HPA antibody, which recognizes the C-domain of HPA inhibited macrophage migration, secretion of IL-1β and TNF-α as well as decreased HPA enzymatic activity and increased Syn-1 protein expression in AGE-induced macrophages. Compared with anti-HPA antibody pretreatment, co-pretreatment with anti-HPA plus Syn-1 antibodies promoted macrophage migration, and secretion of IL-1β and TNF-α significantly in AGE-induced macrophages. In addition, pretreatment with anti-HPA or anti-HPA plus Syn-1 antibodies did not markedly change the mRNA levels of IL-1β and TNF-α concentration AGE-treated macrophages. The results showed that C-domain of HPA mediates AGE-induced macrophage migration and inflammatory cytokine release via Syn-1 protein expression. Furthermore, C-domain of HPA may have a key role in diabetic vascular complication-associated inflammatory response.
Collapse
Affiliation(s)
- Qiaojing Qin
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Guang Xu
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Weiwei Qi
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Mei Guo
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Zhaoxia Wang
- School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Wangjie Xu
- School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Zhongdong Qiao
- School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, P.R. China
| | - Yong Gu
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| | - Jianying Niu
- Department of Nephrology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, P.R. China
| |
Collapse
|
47
|
An X, Zhang L, Yuan Y, Wang B, Yao Q, Li L, Zhang J, He M, Zhang J. Hyperoside pre-treatment prevents glomerular basement membrane damage in diabetic nephropathy by inhibiting podocyte heparanase expression. Sci Rep 2017; 7:6413. [PMID: 28743882 PMCID: PMC5527129 DOI: 10.1038/s41598-017-06844-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/19/2017] [Indexed: 12/22/2022] Open
Abstract
Glomerular basement membrane (GBM) damage plays a pivotal role in pathogenesis of albuminuria in diabetic nephropathy (DN). Heparan sulfate (HS) degradation induced by podocyte heparanase is the major cause of GBM thickening and abnormal perm-selectivity. In the present study, we aimed to examine the prophylactic effect of hyperoside on proteinuria development and GBM damage in DN mouse model and the cultured mouse podocytes. Pre-treatment with hyperoside (30 mg/kg/d) for four weeks could significantly decrease albuminuria, prevent GBM damage and oxidative stress in diabetes mellitus (DM) mice. Immunofluorescence staining, Real time PCR and Western blot analysis showed that decreased HS contents and increased heparanase expression in DN mice were also significantly improved by hyperoside pre-treatment. Meanwhile, transmission electron microscope imaging showed that hyperoside significantly alleviated GBM thickening in DN mice. In addition, hyperoside pre-treatment inhibited the increased heparanase gene (HPR1) promoter activity and heparanase expression induced by high glucose or reactive oxidative species (ROS) in cultured podocytes. Our data suggested that hyperoside has a prophylactic effect on proteinuria development and GBM damage in DM mice by decreasing podocyte heparanase expression.
Collapse
Affiliation(s)
- Xiaofei An
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, 201508, China
| | - Lin Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China
| | - Yanggang Yuan
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province People's Hospital, Nanjing, 210029, China
| | - Bin Wang
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, 201508, China
| | - Qiuming Yao
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, 201508, China
| | - Ling Li
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, 201508, China
| | - Jisheng Zhang
- Department of Otorhinolaryngology, Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - Ming He
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai, 200025, China.
| | - Jinan Zhang
- Department of Endocrinology, Jinshan Hospital of Fudan University, Shanghai, 201508, China.
| |
Collapse
|
48
|
Heparanase Upregulation Contributes to Porcine Reproductive and Respiratory Syndrome Virus Release. J Virol 2017; 91:JVI.00625-17. [PMID: 28490587 DOI: 10.1128/jvi.00625-17] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 05/02/2017] [Indexed: 12/27/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) continues to cause substantial economic losses to the pig industry worldwide. Heparan sulfate (HS) is used by PRRSV for initial attachment to target cells. However, the role of HS in the late phase of PRRSV infection and the mechanism of virus release from host cells remain largely unknown. In this study, we showed that PRRSV infection caused a decrease in HS expression and upregulated heparanase, the only known enzyme capable of degrading HS. We subsequently demonstrated that the NF-κB signaling pathway and cathepsin L protease were involved in regulation of PRRSV infection-induced heparanase. In addition, we found that ablation of heparanase expression using small interfering RNA duplexes increased cell surface expression of HS and suppressed PRRSV replication and release, whereas overexpression of heparanase reduced HS surface expression and enhanced PRRSV replication and release. These data suggest that PRRSV activates NF-κB and cathepsin L to upregulate and process heparanase, and then the active heparanase cleaves HS, resulting in viral release. Our findings provide new insight into the molecular mechanism of PRRSV egress from host cells, which might help us to further understand PRRSV pathogenesis.IMPORTANCE Porcine reproductive and respiratory syndrome virus (PRRSV) causes great economic losses each year to the pig industry worldwide. The molecular mechanism of PRRSV release from host cells largely remains a mystery. In this study, we demonstrate that PRRSV activates NF-κB and cathepsin L to upregulate and process heparanase, and then the active heparanase is released to the extracellular space and exerts enzymatic activity to cleave heparan sulfate, resulting in viral release. Our findings provide new insight into the molecular mechanism of PRRSV egress from host cells, which might help us to further understand PRRSV pathogenesis.
Collapse
|
49
|
Jie L, Pengcheng Q, Qiaoyan H, Linlin B, Meng Z, Fang W, Min J, Li Y, Ya Z, Qian Y, Siwang W. Dencichine ameliorates kidney injury in induced type II diabetic nephropathy via the TGF-β/Smad signalling pathway. Eur J Pharmacol 2017. [PMID: 28633927 DOI: 10.1016/j.ejphar.2017.06.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Diabetic nephropathy (DN), a common complication associated with both type I and type II diabetes mellitus (DM), is a major cause of chronic nephropathy and a common cause of end-stage renal diseases (ESRD) throughout the world. This study is aimed to determine whether dencichine (De) can ameliorate renal damage in high-glucose-and-fat diet combined STZ (streptozocin) induced DN in type II DM rats and to investigate the potential underlying mechanisms. Markers of metabolism, diabetes, and renal function, and levels of extracellular matrix (ECM) collagen I (Col I), collagen IV (Col IV), fibronectin (FN) and laminin (LN), and of proteins in the TGF-β/Smad pathway were analysed through RT-PCR, western blot, immunofluorescence and immunohistochemistry. The results show that De significantly alleviates metabolism disorder, improved renal function, relieved pathological alterations in the glomerulus of DN rats, decreased ECM deposition and increased the ratio of matrix metalloproteinase (MMP)-9 to tissue inhibitor of metalloproteinase (TIMP)-1 both in vivo and in vitro. Moreover, De negatively regulated TGF-β/Smad signalling pathway and increased the expression of Smad7, an endogenic inhibitory Smad located downstream of the signalling pathway. In conclusion, we provide experimental evidence indicating that the renoprotective effect of De could significantly prevent the progression of DN possibly attribute to down-regulation of the TGF-β/Smad pathway and rebalance the deposition and degradation of ECM proteins.
Collapse
Affiliation(s)
- Li Jie
- Department of Natural Medicine, Fourth Military Medical University, 710032 Xi'an, China
| | - Qiu Pengcheng
- Department of Natural Medicine, Fourth Military Medical University, 710032 Xi'an, China
| | - He Qiaoyan
- Department of Natural Medicine, Fourth Military Medical University, 710032 Xi'an, China
| | - Bi Linlin
- Department of Natural Medicine, Fourth Military Medical University, 710032 Xi'an, China
| | - Zhang Meng
- Department of Natural Medicine, Fourth Military Medical University, 710032 Xi'an, China
| | - Wang Fang
- Department of Natural Medicine, Fourth Military Medical University, 710032 Xi'an, China
| | - Jia Min
- Department of Natural Medicine, Fourth Military Medical University, 710032 Xi'an, China; Shaanxi Key Laboratory of Ischemic Cardiovascular Disease, Institute of Basic and Translational Medicine, Xi'an Medical University, 710021 Xi'an, China
| | - Yan Li
- Department of Natural Medicine, Fourth Military Medical University, 710032 Xi'an, China
| | - Zhang Ya
- Department of Natural Medicine, Fourth Military Medical University, 710032 Xi'an, China
| | - Yang Qian
- Department of Natural Medicine, Fourth Military Medical University, 710032 Xi'an, China
| | - Wang Siwang
- Department of Natural Medicine, Fourth Military Medical University, 710032 Xi'an, China.
| |
Collapse
|
50
|
Heparanase: roles in cell survival, extracellular matrix remodelling and the development of kidney disease. Nat Rev Nephrol 2017; 13:201-212. [PMID: 28163306 DOI: 10.1038/nrneph.2017.6] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Heparanase has regulatory roles in various processes, including cell communication, gene transcription and autophagy. In addition, it is the only known mammalian endoglycosidase that is capable of degrading heparan sulfate (HS). HS chains are important constituents and organizers of the extracellular matrix (ECM), and have a key role in maintaining the integrity and function of the glomerular filtration barrier. In addition, HS chains regulate the activity of numerous bioactive molecules, such as cytokines and growth factors, at the cell surface and in the ECM. Given the functional diversity of HS, its degradation by heparanase profoundly affects important pathophysiological processes, including tumour development, neovascularization and inflammation, as well as progression of kidney disease. Heparanase-mediated degradation and subsequent remodelling of HS in the ECM of the glomerulus is a key mechanism in the development of glomerular disease, as exemplified by the complete resistance of heparanase-deficient animals to diabetes and immune-mediated kidney disease. This Review summarizes the role of heparanase in the development of kidney disease, and its potential as a therapeutic target.
Collapse
|