1
|
Darshi M, Kugathasan L, Maity S, Sridhar VS, Fernandez R, Limonte CP, Grajeda BI, Saliba A, Zhang G, Drel VR, Kim JJ, Montellano R, Tumova J, Montemayor D, Wang Z, Liu JJ, Wang J, Perkins BA, Lytvyn Y, Natarajan L, Lim SC, Feldman H, Toto R, Sedor JR, Patel J, Waikar SS, Brown J, Osman Y, He J, Chen J, Reeves WB, de Boer IH, Roy S, Vallon V, Hallan S, Gelfond JA, Cherney DZ, Sharma K. Glycolytic lactate in diabetic kidney disease. JCI Insight 2024; 9:e168825. [PMID: 38855868 PMCID: PMC11382878 DOI: 10.1172/jci.insight.168825] [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: 05/08/2023] [Accepted: 05/01/2024] [Indexed: 06/11/2024] Open
Abstract
Lactate elevation is a well-characterized biomarker of mitochondrial dysfunction, but its role in diabetic kidney disease (DKD) is not well defined. Urine lactate was measured in patients with type 2 diabetes (T2D) in 3 cohorts (HUNT3, SMART2D, CRIC). Urine and plasma lactate were measured during euglycemic and hyperglycemic clamps in participants with type 1 diabetes (T1D). Patients in the HUNT3 cohort with DKD had elevated urine lactate levels compared with age- and sex-matched controls. In patients in the SMART2D and CRIC cohorts, the third tertile of urine lactate/creatinine was associated with more rapid estimated glomerular filtration rate decline, relative to first tertile. Patients with T1D demonstrated a strong association between glucose and lactate in both plasma and urine. Glucose-stimulated lactate likely derives in part from proximal tubular cells, since lactate production was attenuated with sodium-glucose cotransporter-2 (SGLT2) inhibition in kidney sections and in SGLT2-deficient mice. Several glycolytic genes were elevated in human diabetic proximal tubules. Lactate levels above 2.5 mM potently inhibited mitochondrial oxidative phosphorylation in human proximal tubule (HK2) cells. We conclude that increased lactate production under diabetic conditions can contribute to mitochondrial dysfunction and become a feed-forward component to DKD pathogenesis.
Collapse
Affiliation(s)
- Manjula Darshi
- Center for Precision Medicine, Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Luxcia Kugathasan
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, Canada
| | - Soumya Maity
- Center for Precision Medicine, Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Vikas S. Sridhar
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, Canada
| | - Roman Fernandez
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Christine P. Limonte
- Schools of Medicine and Public Health, University of Washington, Seattle, Washington, USA
| | - Brian I. Grajeda
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas, USA
| | - Afaf Saliba
- Center for Precision Medicine, Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Guanshi Zhang
- Center for Precision Medicine, Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Viktor R. Drel
- Center for Precision Medicine, Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Jiwan J. Kim
- Center for Precision Medicine, Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Richard Montellano
- Center for Precision Medicine, Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Jana Tumova
- Center for Precision Medicine, Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Daniel Montemayor
- Center for Precision Medicine, Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Zhu Wang
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Jian-Jun Liu
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | - Jiexun Wang
- Clinical Research Unit, Khoo Teck Puat Hospital, Singapore
| | - Bruce A. Perkins
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, Canada
| | - Yuliya Lytvyn
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, Canada
| | - Loki Natarajan
- Herbert Wertheim School of Public Health, University of California, San Diego, La Jolla, California USA
| | - Su Chi Lim
- Clinical Research Unit & Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore
- Saw Swee Hock School of Public Heath, National University of Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Harold Feldman
- Center for Clinical Epidemiology and Biostatistics and
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Robert Toto
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Texas, USA
| | - John R. Sedor
- Glickman Urology and Kidney and Lerner Research Institutes, Cleveland Clinic, Cleveland, Ohio, USA
| | - Jiten Patel
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Texas, USA
| | - Sushrut S. Waikar
- Section of Nephrology, Boston University School of Medicine and Boston Medical Center, Boston, Massachusetts, USA
| | - Julia Brown
- Division of Nephrology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Yahya Osman
- Division of Nephrology, Department of Medicine, Wayne State University, Detroit, Michigan, USA
| | - Jiang He
- School of Public Health, Tulane University Medical Center, New Orleans, Louisiana, USA
| | - Jing Chen
- Division of Nephrology, Department of Medicine, New Orleans, Louisiana, USA
| | - W. Brian Reeves
- Center for Precision Medicine, Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - Ian H. de Boer
- Schools of Medicine and Public Health, University of Washington, Seattle, Washington, USA
| | - Sourav Roy
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, Texas, USA
| | - Volker Vallon
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
- VA San Diego Healthcare Center, San Diego, California, USA
| | - Stein Hallan
- Department of Clinical and Molecular Medicine, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Nephrology, St. Olav Hospital, Trondheim, Norway
| | - Jonathan A.L. Gelfond
- Department of Population Health Sciences, University of Texas Health San Antonio, San Antonio, Texas, USA
| | - David Z.I. Cherney
- Division of Nephrology, Department of Medicine, University Health Network, Toronto, Canada
| | - Kumar Sharma
- Center for Precision Medicine, Division of Nephrology, Department of Medicine, University of Texas Health San Antonio, San Antonio, Texas, USA
| | | |
Collapse
|
2
|
Mavrogeorgis E, Mischak H, Latosinska A, Vlahou A, Schanstra JP, Siwy J, Jankowski V, Beige J, Jankowski J. Collagen-Derived Peptides in CKD: A Link to Fibrosis. Toxins (Basel) 2021; 14:10. [PMID: 35050988 PMCID: PMC8781252 DOI: 10.3390/toxins14010010] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/07/2021] [Accepted: 12/17/2021] [Indexed: 02/06/2023] Open
Abstract
Collagen is a major component of the extracellular matrix (ECM) and has an imminent role in fibrosis, in, among others, chronic kidney disease (CKD). Collagen alpha-1(I) (col1a1) is the most abundant collagen type and has previously been underlined for its contribution to the disease phenotype. Here, we examined 5000 urinary peptidomic datasets randomly selected from healthy participants or patients with CKD to identify urinary col1a1 fragments and study their abundance, position in the main protein, as well as their correlation with renal function. We identified 707 col1a1 peptides that differed in their amino acid sequence and/or post-translational modifications (hydroxyprolines). Well-correlated peptides with the same amino acid sequence, but a different number of hydroxyprolines, were combined into a final list of 503 peptides. These 503 col1a1 peptides covered 69% of the full col1a1 sequence. Sixty-three col1a1 peptides were significantly and highly positively associated (rho > +0.3) with the estimated glomerular filtration rate (eGFR), while only six peptides showed a significant and strong, negative association (rho < -0.3). A similar tendency was observed for col1a1 peptides associated with ageing, where the abundance of most col1a1 peptides decreased with increasing age. Collectively the results show a strong association between collagen peptides and loss of kidney function and suggest that fibrosis, potentially also of other organs, may be the main consequence of an attenuation of collagen degradation, and not increased synthesis.
Collapse
Affiliation(s)
- Emmanouil Mavrogeorgis
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (E.M.); (H.M.); (A.L.); (J.S.)
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, 52074 Aachen, Germany;
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (E.M.); (H.M.); (A.L.); (J.S.)
| | - Agnieszka Latosinska
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (E.M.); (H.M.); (A.L.); (J.S.)
| | - Antonia Vlahou
- Biotechnology Division, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Joost P. Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1297, Institute of Cardiovascular and Metabolic Disease, 31432 Toulouse, France;
- Université Toulouse III Paul-Sabatier, 31062 Toulouse, France
| | - Justyna Siwy
- Mosaiques Diagnostics GmbH, 30659 Hannover, Germany; (E.M.); (H.M.); (A.L.); (J.S.)
| | - Vera Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, 52074 Aachen, Germany;
| | - Joachim Beige
- Division of Nephrology and KfH Renal Unit, Hospital St Georg, 04129 Leipzig, Germany;
- Department of Internal Medicine 2 (Nephrology, Rheumatology, Endocrinology), Martin-Luther-University Halle/Wittenberg, 06108 Halle/Saale, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University Hospital, 52074 Aachen, Germany;
- Experimental Vascular Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, 6229 Maastricht, The Netherlands
| |
Collapse
|
3
|
Slieker RC, Donnelly LA, Fitipaldi H, Bouland GA, Giordano GN, Åkerlund M, Gerl MJ, Ahlqvist E, Ali A, Dragan I, Elders P, Festa A, Hansen MK, van der Heijden AA, Mansour Aly D, Kim M, Kuznetsov D, Mehl F, Klose C, Simons K, Pavo I, Pullen TJ, Suvitaival T, Wretlind A, Rossing P, Lyssenko V, Legido Quigley C, Groop L, Thorens B, Franks PW, Ibberson M, Rutter GA, Beulens JWJ, 't Hart LM, Pearson ER. Distinct Molecular Signatures of Clinical Clusters in People With Type 2 Diabetes: An IMI-RHAPSODY Study. Diabetes 2021; 70:2683-2693. [PMID: 34376475 PMCID: PMC8564413 DOI: 10.2337/db20-1281] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 08/01/2021] [Indexed: 11/23/2022]
Abstract
Type 2 diabetes is a multifactorial disease with multiple underlying aetiologies. To address this heterogeneity, investigators of a previous study clustered people with diabetes according to five diabetes subtypes. The aim of the current study is to investigate the etiology of these clusters by comparing their molecular signatures. In three independent cohorts, in total 15,940 individuals were clustered based on five clinical characteristics. In a subset, genetic (N = 12,828), metabolomic (N = 2,945), lipidomic (N = 2,593), and proteomic (N = 1,170) data were obtained in plasma. For each data type, each cluster was compared with the other four clusters as the reference. The insulin-resistant cluster showed the most distinct molecular signature, with higher branched-chain amino acid, diacylglycerol, and triacylglycerol levels and aberrant protein levels in plasma were enriched for proteins in the intracellular PI3K/Akt pathway. The obese cluster showed higher levels of cytokines. The mild diabetes cluster with high HDL showed the most beneficial molecular profile with effects opposite of those seen in the insulin-resistant cluster. This study shows that clustering people with type 2 diabetes can identify underlying molecular mechanisms related to pancreatic islets, liver, and adipose tissue metabolism. This provides novel biological insights into the diverse aetiological processes that would not be evident when type 2 diabetes is viewed as a homogeneous disease.
Collapse
Affiliation(s)
- Roderick C Slieker
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Epidemiology and Data Science, Amsterdam Public Health Institute, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
| | - Louise A Donnelly
- Population Health & Genomics, School of Medicine, University of Dundee, Dundee, U.K
| | - Hugo Fitipaldi
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, SUS, Malmö, Sweden
| | - Gerard A Bouland
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Giuseppe N Giordano
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, SUS, Malmö, Sweden
| | - Mikael Åkerlund
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, SUS, Malmö, Sweden
| | | | - Emma Ahlqvist
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, SUS, Malmö, Sweden
| | - Ashfaq Ali
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Iulian Dragan
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Petra Elders
- Department of General Practice and Elderly Care Medicine, Amsterdam Public Health Research Institute, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
| | - Andreas Festa
- Eli Lilly Regional Operations GmbH, Vienna, Austria
- 1st Medical Department, LK Stockerau, Niederösterreich, Austria
| | - Michael K Hansen
- Cardiovascular and Metabolic Disease Research, Janssen Research & Development, Spring House, PA
| | - Amber A van der Heijden
- Department of General Practice and Elderly Care Medicine, Amsterdam Public Health Research Institute, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
| | - Dina Mansour Aly
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, SUS, Malmö, Sweden
| | - Min Kim
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Institute of Pharmaceutical Science, Faculty of Life Sciences and Medicines, King's College London, London, U.K
| | - Dmitry Kuznetsov
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Florence Mehl
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | | | - Imre Pavo
- Eli Lilly Regional Operations GmbH, Vienna, Austria
| | - Timothy J Pullen
- Department of Diabetes, Guy's Campus, King's College London, London, U.K
- Section of Cell Biology and Functional Genomics, Division of Diabetes, Endocrinology and Metabolism, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, U.K
| | | | | | - Peter Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Valeriya Lyssenko
- Department of Clinical Science, Center for Diabetes Research, University of Bergen, Bergen, Norway
- Genomics, Diabetes and Endocrinology Unit, Department of Clinical Sciences Malmö, Lund University Diabetes Centre, Skåne University Hospital, Malmö, Sweden
| | - Cristina Legido Quigley
- Steno Diabetes Center Copenhagen, Gentofte, Denmark
- Cardiovascular and Metabolic Disease Research, Janssen Research & Development, Spring House, PA
| | - Leif Groop
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, SUS, Malmö, Sweden
- Finnish Institute of Molecular Medicine, Helsinki University, Helsinki, Finland
| | - Bernard Thorens
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Paul W Franks
- Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, SUS, Malmö, Sweden
- Department of Nutrition, Harvard School of Public Health, Boston, MA
| | - Mark Ibberson
- Vital-IT Group, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Guy A Rutter
- Department of Diabetes, Guy's Campus, King's College London, London, U.K
- Lee Kong Chian School of Medicine, Nan Yang Technological University, Singapore
| | - Joline W J Beulens
- Department of Epidemiology and Data Science, Amsterdam Public Health Institute, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Leen M 't Hart
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
- Department of Epidemiology and Data Science, Amsterdam Public Health Institute, Amsterdam UMC, location VUmc, Amsterdam, the Netherlands
- Molecular Epidemiology, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Ewan R Pearson
- Population Health & Genomics, School of Medicine, University of Dundee, Dundee, U.K.
| |
Collapse
|
4
|
Kong W, Lyu C, Liao H, Du Y. Collagen crosslinking: effect on structure, mechanics and fibrosis progression. Biomed Mater 2021; 16. [PMID: 34587604 DOI: 10.1088/1748-605x/ac2b79] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 09/29/2021] [Indexed: 02/07/2023]
Abstract
Biophysical properties of extracellular matrix (ECM), such as matrix stiffness, viscoelasticity and matrix fibrous structure, are emerging as important factors that regulate progression of fibrosis and other chronic diseases. The biophysical properties of the ECM can be rapidly and profoundly regulated by crosslinking reactions in enzymatic or non-enzymatic manners, which further alter the cellular responses and drive disease progression. In-depth understandings of crosslinking reactions will be helpful to reveal the underlying mechanisms of fibrosis progression and put forward new therapeutic targets, whereas related reviews are still devoid. Here, we focus on the main crosslinking mechanisms that commonly exist in a plethora of chronic diseases (e.g. fibrosis, cancer, osteoarthritis) and summarize current understandings including the biochemical reaction, the effect on ECM properties, the influence on cellular behaviors, and related studies in disease model establishment. Potential pharmaceutical interventions targeting the crosslinking process and relevant clinical studies are also introduced. Limitations of pharmaceutical development may be due to the lack of systemic investigations related to the influence on crosslinking mechanism from micro to macro level, which are discussed in the last section. We also propose the unclarified questions regarding crosslinking mechanisms and potential challenges in crosslinking-targeted therapeutics development.
Collapse
Affiliation(s)
- Wenyu Kong
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, People's Republic of China
| | - Cheng Lyu
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, People's Republic of China
| | - Hongen Liao
- Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing 100084, People's Republic of China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing 100084, People's Republic of China
| |
Collapse
|
5
|
Bódi N, Mezei D, Chakraborty P, Szalai Z, Barta BP, Balázs J, Rázga Z, Hermesz E, Bagyánszki M. Diabetes-related intestinal region-specific thickening of ganglionic basement membrane and regionally decreased matrix metalloproteinase 9 expression in myenteric ganglia. World J Diabetes 2021; 12:658-672. [PMID: 33995853 PMCID: PMC8107976 DOI: 10.4239/wjd.v12.i5.658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/10/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The importance of the neuronal microenvironment has been recently highlighted in gut region-specific diabetic enteric neuropathy. Regionally distinct thickening of endothelial basement membrane (BM) of intestinal capillaries supplying the myenteric ganglia coincide with neuronal damage in different intestinal segments. Accelerated synthesis of matrix molecules and reduced degradation of matrix components may also contribute to the imbalance of extracellular matrix dynamics resulting in BM thickening. Among the matrix degrading proteinases, matrix metalloproteinase 9 (MMP9) and its tissue inhibitor (TIMP1) are essential in regulating extracellular matrix remodelling.
AIM To evaluate the intestinal segment-specific effects of diabetes and insulin replacement on ganglionic BM thickness, MMP9 and TIMP1 expression.
METHODS Ten weeks after the onset of hyperglycaemia gut segments were taken from the duodenum and ileum of streptozotocin-induced diabetic, insulin-treated diabetic and sex- and age-matched control rats. The thickness of BM surrounding myenteric ganglia was measured by electron microscopic morphometry. Whole-mount preparations of myenteric plexus were prepared from the different gut regions for MMP9/TIMP1 double-labelling fluorescent immunohistochemistry. Post-embedding immunogold electron microscopy was applied on ultrathin sections to evaluate the MMP9 and TIMP1 expression in myenteric ganglia and their microenvironment from different gut segments and conditions. The MMP9 and TIMP1 messenger ribonucleic acid (mRNA) level was measured by quantitative polymerase chain reaction.
RESULTS Ten weeks after the onset of hyperglycaemia, the ganglionic BM was significantly thickened in the diabetic ileum, while it remained intact in the duodenum. The immediate insulin treatment prevented the diabetes-related thickening of the BM surrounding the ileal myenteric ganglia. Quantification of particle density showed an increasing tendency for MMP9 and a decreasing tendency for TIMP1 from the proximal to the distal small intestine under control conditions. In the diabetic ileum, the number of MMP9-indicating gold particles decreased in myenteric ganglia, endothelial cells of capillaries and intestinal smooth muscle cells, however, it remained unchanged in all duodenal compartments. The MMP9/TIMP1 ratio was also decreased in ileal ganglia only. However, a marked segment-specific induction was revealed in MMP9 and TIMP1 at the mRNA levels.
CONCLUSION These findings support that the regional decrease in MMP9 expression in myenteric ganglia and their microenvironment may contribute to extracellular matrix accumulation, resulting in a region-specific thickening of ganglionic BM.
Collapse
Affiliation(s)
- Nikolett Bódi
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - Diána Mezei
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - Payal Chakraborty
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - Zita Szalai
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - Bence Pál Barta
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - János Balázs
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - Zsolt Rázga
- Department of Pathology, Faculty of Medicine, University of Szeged, Szeged 6720, Hungary
| | - Edit Hermesz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| | - Mária Bagyánszki
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged 6726, Hungary
| |
Collapse
|
6
|
Matrix Metalloproteinases in Diabetic Kidney Disease. J Clin Med 2020; 9:jcm9020472. [PMID: 32046355 PMCID: PMC7073625 DOI: 10.3390/jcm9020472] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 12/11/2022] Open
Abstract
Around the world diabetic kidney disease (DKD) is the main cause of chronic kidney disease (CKD), which is characterized by mesangial expansion, glomerulosclerosis, tubular atrophy, and interstitial fibrosis. The hallmark of the pathogenesis of DKD is an increased extracellular matrix (ECM) accumulation causing thickening of the glomerular and tubular basement membranes, mesangial expansion, sclerosis, and tubulointerstitial fibrosis. The matrix metalloproteases (MMPs) family are composed of zinc-dependent enzymes involved in the degradation and hydrolysis of ECM components. Several MMPs are expressed in the kidney; nephron compartments, vasculature and connective tissue. Given their important role in DKD, several studies have been performed in patients with DKD proposing that the measurement of their activity in serum or in urine may become in the future markers of early DKD. Studies from diabetic nephropathy experimental models suggest that a balance between MMPs levels and their inhibitors is needed to maintain renal homeostasis. This review focuses in the importance of the MMPs within the kidney and their modifications at the circulation, kidney and urine in patients with DKD. We also cover the most important studies performed in experimental models of diabetes in terms of MMPs levels, renal expression and its down-regulation effect.
Collapse
|
7
|
Sugahara M, Tanaka S, Tanaka T, Saito H, Ishimoto Y, Wakashima T, Ueda M, Fukui K, Shimizu A, Inagi R, Yamauchi T, Kadowaki T, Nangaku M. Prolyl Hydroxylase Domain Inhibitor Protects against Metabolic Disorders and Associated Kidney Disease in Obese Type 2 Diabetic Mice. J Am Soc Nephrol 2020; 31:560-577. [PMID: 31996409 DOI: 10.1681/asn.2019060582] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 12/12/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Prolyl hydroxylase domain (PHD) inhibitors, which stimulate erythropoietin production through the activation of hypoxia-inducible factor (HIF), are novel therapeutic agents used for treating renal anemia. Several PHD inhibitors, including enarodustat, are currently undergoing phase 2 or phase 3 clinical trials. Because HIF regulates a broad spectrum of genes, PHD inhibitors are expected to have other effects in addition to erythropoiesis, such as protection against metabolic disorders. However, whether such beneficial effects would extend to metabolic disorder-related kidney disease is largely unknown. METHODS We administered enarodustat or vehicle without enarodustat in feed to diabetic black and tan brachyury (BTBR) ob/ob mice from 4 to 22 weeks of age. To elucidate molecular changes induced by enarodustat, we performed transcriptome analysis of isolated glomeruli and in vitro experiments using murine mesangial cells. RESULTS Compared with BTBR ob/ob mice that received only vehicle, BTBR ob/ob mice treated with enarodustat displayed lower body weight, reduced blood glucose levels with improved insulin sensitivity, lower total cholesterol levels, higher adiponectin levels, and less adipose tissue, as well as a tendency for lower macrophage infiltration. Enarodustat-treated mice also exhibited reduced albuminuria and amelioration of glomerular epithelial and endothelial damage. Transcriptome analysis of isolated glomeruli revealed reduced expression of C-C motif chemokine ligand 2/monocyte chemoattractant protein-1 (CCL2/MCP-1) in enarodustat-treated mice compared with the vehicle-only group, accompanied by reduced glomerular macrophage infiltration. In vitro experiments demonstrated that both local HIF-1 activation and restoration of adiponectin by enarodustat contributed to CCL2/MCP-1 reduction in mesangial cells. CONCLUSIONS These results indicate that the PHD inhibitor enarodustat has potential renoprotective effects in addition to its potential to protect against metabolic disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Takeshi Wakashima
- Biological and Pharmacological Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan; and
| | - Masatoshi Ueda
- Biological and Pharmacological Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan; and
| | - Kenji Fukui
- Biological and Pharmacological Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan; and
| | - Akira Shimizu
- Department of Analytic Human Pathology, Nippon Medical School, Tokyo, Japan
| | - Reiko Inagi
- Division of Chronic Kidney Disease Pathophysiology, and
| | - Toshimasa Yamauchi
- Department of Diabetes and Metabolic Diseases, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | - Takashi Kadowaki
- Department of Diabetes and Metabolic Diseases, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| | | |
Collapse
|
8
|
Poteryaeva ON, Usynin IF. [Antidiabetic role of high density lipoproteins]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2019; 64:463-471. [PMID: 30632974 DOI: 10.18097/pbmc20186406463] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Disturbance in lipid metabolism can be both a cause and a consequence of the development of diabetes mellitus (DM). One of the most informative indicator of lipid metabolism is the ratio of atherogenic and antiatherogenic fractions of lipoproteins and their protein components. The review summarizes literature data and own results indicating the important role of high-density lipoprotein (HDL) and their main protein component, apolipoprotein A-I (apoA-I), in the pathogenesis of type 2 DM. On the one hand, HDL are involved in the regulation of insulin secretion by b-cells and insulin-independent absorption of glucose. On the other hand, insulin resistance and hyperglycemia lead to a decrease in HDL levels and cause modification of their protein component. In addition, HDL, possessing anti-inflammatory and mitogenic properties, provide anti-diabetic protection through systemic mechanisms. Thus, maintaining a high concentration of HDL and apoA-I in blood plasma and preventing their modification are important issues in the context of prevention and treatment of diabetes.
Collapse
Affiliation(s)
- O N Poteryaeva
- Institute of Biochemistry, Federal Research Center of Fundamental and Translation Medicine, Novosibirsk, Russia
| | - I F Usynin
- Institute of Biochemistry, Federal Research Center of Fundamental and Translation Medicine, Novosibirsk, Russia
| |
Collapse
|
9
|
Chrysin Inhibits Advanced Glycation End Products-Induced Kidney Fibrosis in Renal Mesangial Cells and Diabetic Kidneys. Nutrients 2018; 10:nu10070882. [PMID: 29987200 PMCID: PMC6073220 DOI: 10.3390/nu10070882] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 06/30/2018] [Accepted: 07/06/2018] [Indexed: 12/13/2022] Open
Abstract
Advanced glycation end products (AGEs) play a causative role in the development of diabetic nephropathy via induction of matrix protein deposition in kidneys. This study investigated inhibitory effects of chrysin, present in bee propolis and herbs, on glomerulosclerosis in db/db mice and AGEs-exposed renal mesangial cells. The in vivo study explored the demoting effects of 10 mg/kg chrysin on glomerular fibrosis in a type 2 diabetic model. Oral supplementation of chrysin inhibited the collagen fiber accumulation and α-smooth muscle actin (α-SMA) induction in periodic acid schiff-positive renal tissues of db/db mice. Moreover, treating db/db mice with chrysin diminished the level of AGEs increased in diabetic glomeruli. The in vitro study employed human mesangial cells exposed to 100 μg/mL AGE-BSA for 72 h in the presence of 1⁻20 μM chrysin. Glucose increased mesangial AGE production via induction of receptor for AGEs. Chrysin suppressed the induction of collagens, α-SMA, fibroblast-specific protein-1 and matrix metalloproteinases enhanced by AGE-bovine serum albumin. Furthermore, chrysin blunted transforming growth factor-β1 induction and Smad 2/3 activation in AGEs-exposed mesangial cells. These results demonstrate that chrysin attenuated accumulation of myofibroblast-like cells and matrix proteins in AGEs-laden diabetic glomeruli. Therefore, chrysin may be a potential renoprotective agent targeting glucose-mediated AGEs-associated glomerulosclerosis and fibrosis.
Collapse
|
10
|
Zeidán-Chuliá F, Yilmaz D, Häkkinen L, Könönen E, Neves de Oliveira BH, Güncü G, Uitto VJ, Caglayan F, Gürsoy UK. Matrix metalloproteinase-7 in periodontitis with type 2 diabetes mellitus. J Periodontal Res 2018; 53:916-923. [DOI: 10.1111/jre.12583] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/27/2018] [Indexed: 12/24/2022]
Affiliation(s)
- F. Zeidán-Chuliá
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica; Departamento de Bioquímica; Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul (UFRGS); Porto Alegre RS Brazil
- Department of Periodontology; Institute of Dentistry; University of Turku; Turku Finland
- Departamento de Ciencias Biomédicas Básicas; Facultad de Ciencias Biomédicas y de la Salud; Universidad Europea de Madrid; Villaviciosa de Odón Spain
| | - D. Yilmaz
- Department of Periodontology; Institute of Dentistry; University of Turku; Turku Finland
- Department of Periodontology; Faculty of Dentistry; University of Sakarya; Sakarya Turkey
| | - L. Häkkinen
- Laboratory of Periodontal Biology; Faculty of Dentistry; University of British Columbia; Vancouver BC Canada
| | - E. Könönen
- Department of Periodontology; Institute of Dentistry; University of Turku; Turku Finland
- Oral Health Care; Welfare Division; City of Turku; Turku Finland
| | - B.-H. Neves de Oliveira
- Programa de Pós-Graduação em Ciências Biológicas, Bioquímica; Departamento de Bioquímica; Instituto de Ciências Básicas da Saúde; Universidade Federal do Rio Grande do Sul (UFRGS); Porto Alegre RS Brazil
| | - G. Güncü
- Department of Periodontology; Faculty of Dentistry; University of Hacettepe; Ankara Turkey
| | - V.-J. Uitto
- Department of Oral Biology; Institute of Dentistry; University of Helsinki; Helsinki Finland
| | - F. Caglayan
- Department of Periodontology; Faculty of Dentistry; University of Hacettepe; Ankara Turkey
| | - U. K. Gürsoy
- Department of Periodontology; Institute of Dentistry; University of Turku; Turku Finland
| |
Collapse
|
11
|
Poteryaeva ON, Russkikh GS, Zubova AV, Gevorgyan MM, Usynin IF. Changes in Activity of Matrix Metalloproteinases and Serum Concentrations of Proinsulin and C-Peptide Depending on the Compensation Stage of Type 2 Diabetes Mellitus. Bull Exp Biol Med 2018; 164:730-733. [DOI: 10.1007/s10517-018-4068-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Indexed: 12/12/2022]
|
12
|
Peeters SA, Engelen L, Buijs J, Theilade S, Rossing P, Schalkwijk CG, Stehouwer CDA. Associations between advanced glycation endproducts and matrix metalloproteinases and its inhibitor in individuals with type 1 diabetes. J Diabetes Complications 2018; 32:325-329. [PMID: 29395841 DOI: 10.1016/j.jdiacomp.2017.12.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/30/2017] [Accepted: 12/26/2017] [Indexed: 12/14/2022]
Abstract
AIMS Advanced glycation endproducts (AGEs) and altered extracellular matrix remodeling by matrix metalloproteinases (MMPs) and tissue inhibitor of metalloproteinase (TIMP) are associated with vascular complications in type 1 diabetes. Experimental studies have shown that AGEs regulate the production of MMPs and/or TIMP-1. Therefore, we investigated associations between specific AGEs and MMP-1, -2, -3, -9, and -10, and TIMP-1 in individuals with type 1 diabetes. METHODS In 670 type 1 diabetic individuals we determined serum levels of protein-bound AGEs Nε-(carboxymethyl)lysine (CML), Nε-(carboxyethyl)lysine (CEL), 5-hydro-5-methylimidazolone (MG-H1) and pentosidine, and MMP-1, -2, -3, -9, and -10, and TIMP-1. We performed linear regression analyses to investigate associations between AGEs and markers of the MMP-TIMP system. Analyses were adjusted for age, sex, HbA1c and duration of diabetes, and additionally for other potential confounders and presence of vascular complication. RESULTS After full adjustment, levels of CML were positively associated with levels of MMP-2 and inversely with MMP-9. CEL was positively associated with MMP-3 and TIMP-1. MG-H1 was only associated with TIMP-1, whereas pentosidine was not associated with MMPs or TIMP-1. CONCLUSIONS We showed independent associations between several AGEs and markers of the MMP-TIMP system, which indicate specific AGE-MMP/TIMP-1 interactions potentially contributing to vascular complications in patients with type 1 diabetes.
Collapse
Affiliation(s)
- S A Peeters
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands; Department of Internal Medicine, Zuyderland hospital, Heerlen, The Netherlands.
| | - L Engelen
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands; Centraal Bureau voor de Statistiek, Heerlen, The Netherlands
| | - J Buijs
- Department of Internal Medicine, Zuyderland hospital, Heerlen, The Netherlands.
| | - S Theilade
- Steno Diabetes Center Copenhagen, Gentofte, Denmark.
| | - P Rossing
- Steno Diabetes Center Copenhagen, Gentofte, Denmark; University of Copenhagen, Copenhagen, Denmark.
| | - C G Schalkwijk
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands.
| | - C D A Stehouwer
- Department of Internal Medicine, Maastricht University Medical Centre, Maastricht, The Netherlands.
| |
Collapse
|
13
|
Krochmal M, Kontostathi G, Magalhães P, Makridakis M, Klein J, Husi H, Leierer J, Mayer G, Bascands JL, Denis C, Zoidakis J, Zürbig P, Delles C, Schanstra JP, Mischak H, Vlahou A. Urinary peptidomics analysis reveals proteases involved in diabetic nephropathy. Sci Rep 2017; 7:15160. [PMID: 29123184 PMCID: PMC5680307 DOI: 10.1038/s41598-017-15359-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022] Open
Abstract
Mechanisms underlying the onset and progression of nephropathy in diabetic patients are not fully elucidated. Deregulation of proteolytic systems is a known path leading to disease manifestation, therefore we hypothesized that proteases aberrantly expressed in diabetic nephropathy (DN) may be involved in the generation of DN-associated peptides in urine. We compared urinary peptide profiles of DN patients (macroalbuminuric, n = 121) to diabetic patients with no evidence of DN (normoalbuminuric, n = 118). 302 sequenced, differentially expressed peptides (adjusted p-value < 0.05) were analysed with the Proteasix tool predicting proteases potentially involved in their generation. Activity change was estimated based on the change in abundance of the investigated peptides. Predictions were correlated with transcriptomics (Nephroseq) and relevant protein expression data from the literature. This analysis yielded seventeen proteases, including multiple forms of MMPs, cathepsin D and K, kallikrein 4 and proprotein convertases. The activity of MMP-2 and MMP-9, predicted to be decreased in DN, was investigated using zymography in a DN mouse model confirming the predictions. Collectively, this proof-of-concept study links urine peptidomics to molecular changes at the tissue level, building hypotheses for further investigation in DN and providing a workflow with potential applications to other diseases.
Collapse
Affiliation(s)
| | | | - Pedro Magalhães
- Mosaiques Diagnostics GmbH, Hannover, Germany
- Department of Pediatric Nephrology, Hannover Medical School, Hannover, Germany
| | | | - Julie Klein
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Holger Husi
- Department of Diabetes and Cardiovascular Science, University of the Highlands and Islands, Centre for Health Science, Inverness, IV2 3JH, UK
| | - Johannes Leierer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| | - Gert Mayer
- Department of Internal Medicine IV (Nephrology and Hypertension), Medical University Innsbruck, Innsbruck, Austria
| | - Jean-Loup Bascands
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1188 - Université de La, Réunion, France
| | - Colette Denis
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Jerome Zoidakis
- Biomedical Research Foundation Academy of Athens, Athens, Greece
| | | | - Christian Delles
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France
- Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Harald Mischak
- Mosaiques Diagnostics GmbH, Hannover, Germany
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, 126 University Place, Glasgow, G12 8TA, UK
| | - Antonia Vlahou
- Biomedical Research Foundation Academy of Athens, Athens, Greece.
| |
Collapse
|
14
|
Zhao Y, Shen Z, Zhang D, Luo H, Chen J, Sun Y, Xiao Q. Ghrelin ameliorates nerve growth factor Dysmetabolism and inflammation in STZ-induced diabetic rats. Metab Brain Dis 2017; 32:903-912. [PMID: 28357639 DOI: 10.1007/s11011-017-0001-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 03/23/2017] [Indexed: 01/07/2023]
Abstract
Diabetic encephalopathy is characterized by cognitive impairment and neuroinflammation, deficient neurotrophic support, and neuronal and synaptic loss. Ghrelin, a 28 amino acid peptide, is associated with neuromodulation and cognitive improvement, which has been considered as a potential protective agent for several neurodegenerative diseases. Here we sought to investigate the role of ghrelin in preventing diabetic-related neuropathology. We found that ghrelin attenuated astrocytic activation and reduced levels of interleukin-6 and tumor necrosis factor-α in streptozotocin-induced diabetic rats. In addition, ghrelin inhibited p38 mitogen-associated protein kinase activation. The upregulation of nerve growth factor (NGF) precursor and matrix metalloproteinase (MMP)-9 and downregulation of mature NGF and MMP-7 in the diabetic brain were reversed by ghrelin. Treatment with ghrelin elevated synaptophysin expression and synaptic density in diabetic rats. Taken together, our results demonstrate that ghrelin ameliorates diabetes-related neurodegeneration by preventing NGF dysmetabolism and synaptic degeneration through regulating MMP levels as well as inhibiting neuroinflammation.
Collapse
Affiliation(s)
- Yuxing Zhao
- Department of Geriatrics, the First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, YuZhong District, Chongqing, 400016, China
| | - Zhaoxing Shen
- Department of Geriatrics, the First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, YuZhong District, Chongqing, 400016, China
| | - Dongling Zhang
- Department of Geriatrics, the First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, YuZhong District, Chongqing, 400016, China
| | - Huiqiong Luo
- Department of Geriatrics, the First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, YuZhong District, Chongqing, 400016, China
| | - Jinliang Chen
- Department of Geriatrics, the First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, YuZhong District, Chongqing, 400016, China
| | - Yue Sun
- Department of Geriatrics, the First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, YuZhong District, Chongqing, 400016, China
| | - Qian Xiao
- Department of Geriatrics, the First Affiliated Hospital of Chongqing Medical University, No. 1 YouYi Road, YuZhong District, Chongqing, 400016, China.
| |
Collapse
|
15
|
Parrish AR. Matrix Metalloproteinases in Kidney Disease: Role in Pathogenesis and Potential as a Therapeutic Target. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:31-65. [PMID: 28662825 DOI: 10.1016/bs.pmbts.2017.03.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Matrix metalloproteinases (MMPs) are large family of proteinases. In addition to a fundamental role in the remodeling of the extracellular matrix, they also cleave a number of cell surface proteins and are involved in multiple cellular processes. MMP activity is regulated via numerous mechanisms, including inhibition by endogenous tissue inhibitors of metalloproteinases (TIMPs). Similar to MMPs, a role for TIMPs has been established in multiple cell signaling pathways. Aberrant expression of MMPs and TIMPS in renal pathophysiology has long been recognized, and with the generation of specific knockout mice, the mechanistic role of several MMPs and TIMPs is becoming more understood and has revealed both pathogenic and protective roles. This chapter will focus on the expression and localization of MMPs and TIMPs in the kidney, as well as summarizing the current information linking these proteins to acute kidney injury and chronic kidney disease. In addition, we will summarize studies suggesting that MMPs and TIMPs may be biomarkers of renal dysfunction and represent novel therapeutic targets to attenuate kidney disease.
Collapse
Affiliation(s)
- Alan R Parrish
- School of Medicine, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
16
|
Ke B, Fan C, Yang L, Fang X. Matrix Metalloproteinases-7 and Kidney Fibrosis. Front Physiol 2017; 8:21. [PMID: 28239354 PMCID: PMC5301013 DOI: 10.3389/fphys.2017.00021] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/10/2017] [Indexed: 12/18/2022] Open
Abstract
Matrix metalloproteinase-7 (MMP-7) is a secreted zinc- and calcium-dependent endopeptidase that degrades a broad range of extracellular matrix substrates and additional substrates. MMP-7 playsa crucial role in a diverse array of cellular processes and appears to be a key regulator of fibrosis in several diseases, including pulmonary fibrosis, liver fibrosis, and cystic fibrosis. In particular, the relationship between MMP-7 and kidney fibrosis has attracted significant attention in recent years. Growing evidence indicates that MMP-7 plays an important role in the pathogenesis of kidney fibrosis. Here, we summarize the recent progress in the understanding of the role of MMP-7 in kidney fibrosis. In particular, we discuss how MMP-7 contributes to kidney fibrotic lesions via the following three pathways: epithelial-mesenchymal transition (EMT), transforming growth factor-beta (TGF-β) signaling, and extracellular matrix (ECM) deposition. Further dissection of the crosstalk among and regulation of these pathways will help clinicians and researchers develop effective therapeutic approaches for treating chronic kidney disease.
Collapse
Affiliation(s)
- Ben Ke
- The Third Hospital of Nanchang Nanchang, China
| | - Chuqiao Fan
- Nanchang University School of Medicine Nanchang, China
| | - Liping Yang
- Department of Nephrology, The Second Affiliated Hospital to Nanchang University Nanchang, China
| | - Xiangdong Fang
- Department of Breast Surgery, Jiangxi Cancer Hospital Nanchang, China
| |
Collapse
|
17
|
Fang J, Wang W, Sun S, Wang Y, Li Q, Lu X, Qiu M, Zhang Y. Metabolomics study of renal fibrosis and intervention effects of total aglycone extracts of Scutellaria baicalensis in unilateral ureteral obstruction rats. JOURNAL OF ETHNOPHARMACOLOGY 2016; 192:20-29. [PMID: 27286917 DOI: 10.1016/j.jep.2016.06.014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 04/09/2016] [Accepted: 06/04/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Scutellariae Radix (Scutellaria baicalensis Georgi) is a well-known Traditional Chinese Medicine (TCM) which mainly contains flavonoids. Our previous studies have demonstrated that total aglycone extracts of Scutellaria baicalensis (TAES) can improve kidney disease in rats. AIM OF THE STUDY To investigate the renal fibrosis (RF) pathogenesis and TAES treatment mechanism in unilateral ureteral obstruction (UUO) rats, using a metabolomics approach based on gas chromatography-mass spectrometry (GC/MS). METHODS Rats with RF were divided into 6 groups with rats subjected to sham operation as normal control. The effects of TAES on some RF closely related parameters in UUO rats were investigated. A metabolomics method, based on GC/MS, was developed to monitor metabolic alterations in urine. Multivariate data analysis was utilized to identify biomarkers potentially associated with RF and the anti-RF activity of TAES. Ontology-based enrichment analysis by BiNChE and pathway analysis by MetPA aid in the interpretation of difference metabolites. RESULTS After 10 days of treatment, the parameters of renal function begin returning to normal, and the abnormal high expressions of genes associated with extracellular matrix (ECM) were relived. In the metabolomics study, metabolic perturbations induced by UUO were reversed after treatment and TAES showed a dose-dependent therapy effect on RF, meanwhile, 18 potential biomarkers associated with RF were identified. Enrichment analysis of metabolites shows an over representation of mostly alkane-alpha, omega-diamine and alpha, omega-dicarboxylic acid, and these biomarkers are primarily involved in Glycine, serine and threonine metabolism, Retinol metabolism, Arginine and proline metabolism and Fructose and mannose metabolism. CONCLUSIONS Our findings indicate that TAES have positive effects on UUO-induced RF in rats, meanwhile, metabolomics method coupled with metabolites enrichment analysis is a useful tool for revealing the pathogenesis of diseases and action mechanism of TCM on the whole body.
Collapse
Affiliation(s)
- Junwei Fang
- Center for Traditional Chinese Medicine and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenyu Wang
- MacroStat (China) Clinical Research Co., Ltd, Shanghai 201203, China
| | - Shujun Sun
- Center for Traditional Chinese Medicine and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yang Wang
- Center for Traditional Chinese Medicine and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qianhua Li
- Center for Traditional Chinese Medicine and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiong Lu
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Mingfeng Qiu
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Yongyu Zhang
- Center for Traditional Chinese Medicine and Systems Biology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
18
|
Tipton DA, Hatten AA, Babu JP, Dabbous MK. Effect of glycated albumin and cranberry components on interleukin-6 and matrix metalloproteinase-3 production by human gingival fibroblasts. J Periodontal Res 2015; 51:228-36. [PMID: 26179241 DOI: 10.1111/jre.12302] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/17/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND OBJECTIVE Gingival fibroblasts have the potential to participate in periodontal inflammation and breakdown, producing interleukin (IL)-6 and matrix metalloproteinase (MMP)-3. Advanced glycation end products (AGEs), formed during diabetic hyperglycemia, might aggravate periodontal inflammation. The cranberry contains anti-inflammatory polyphenols, which inhibit proinflammatory activities of lipopolysaccharide (LPS)- and IL-1β-stimulated human cells. Little is known of its effects on gingival fibroblast IL-6 or MMP-3 production stimulated by AGEs. The objectives were to determine cranberry effects on IL-6 and MMP-3 production by gingival fibroblasts exposed to the representative AGE, glycated human serum albumin (G-HSA), or LPS ± G-HSA. MATERIAL AND METHODS Cranberry high molecular weight non-dialyzable material (NDM), was derived from cranberry juice. Normal human gingival fibroblasts were incubated with G-HSA or normal HSA or Porphyromonas gingivalis LPS (1 μg/mL) ± G-HSA, in the presence or absence of preincubation with NDM. IL-6 and MMP-3 were measured by enzyme-linked immunosorbent assay. Data were analyzed using one-way analysis of variance and Scheffe's F procedure. RESULTS IL-6 production was stimulated by G-HSA or LPS (p < 0.01), which was inhibited in both cases by NDM (p < 0.002). [G-HSA+LPS] synergistically stimulated IL-6 production (p < 0.0001), which was inhibited by NDM. MMP-3 levels were not stimulated by G-HSA but were decreased by LPS (p < 0.02). [G-HSA+LPS] increased MMP-3 production significantly, vs. LPS (p = 0.0005). NDM inhibited MMP-3 levels in the presence of G-HSA or LPS, and in the presence of [G-HSA+LPS] (p < 0.0001). CONCLUSIONS G-HSA ± LPS may have differential effects on IL-6 and MMP-3 production by human gingival fibroblasts, but both are inhibited by NDM. The study suggests that cranberry phenols may be useful in regulating the host response and perhaps treating periodontitis in patients with poorly controlled diabetes.
Collapse
Affiliation(s)
- D A Tipton
- College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - A A Hatten
- College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - J P Babu
- College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - M Kh Dabbous
- College of Dentistry, University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA.,College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA.,Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
19
|
NIRALA BIKESHK, GOHIL NIVEDITAKARMAKAR. EFFECT OF GLYCATED SERUM ALBUMIN ON FUNCTIONAL MARKERS IN HUMAN UMBILICAL VEIN ENDOTHELIAL CELLS IN THE PRESENCE OF SHEAR STRESS. J MECH MED BIOL 2015. [DOI: 10.1142/s0219519415500268] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Regions of the vasculature subjected to low wall shear stress and disturbed flow patterns are prone to atherosclerotic plaque formation. Pro-inflammatory conditions induced by products of protein glycation in diabetes substantially enhance this risk. One of the contributory factors is the enhanced production of ROS by advanced glycation end products (AGE) of proteins and lipids formed in chronic diabetes. In this study, we examine the interaction of oscillatory wall shear stress and glycated serum albumin (AGE-HSA) in modulating NOsynthase activity and expression of pro-inflammatory molecules such as RAGE, s-ICAM-1 and matrix metalloproteinase (MMP-9) in cultured HUVEC. Our findings indicate that orbital shear stress (OSS) up-regulates RAGE expression at low (LSS 4.5 dyn/cm2) more than at high shear stress (HSS 12 dyn/cm2) at 4 h. This effect is promoted in the AGE-HSA (2 mg/mL) in a dose-dependent manner. Augmentation of NOsynthase activity was lower at LSS and further inhibited in the presence of AGE-HSA. Expression of s-ICAM-1 was found to be shear stress modulated with additive up-regulation in combination with AGE-HSA while MMP-9 was not affected by shear stress or AGE-HSA individually but in combination caused significant up-regulation. These changes in endothelial function correlate with mechanisms that initiate atherogenic process in diabetic macrovascular pathology.
Collapse
Affiliation(s)
- BIKESH K. NIRALA
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - NIVEDITA KARMAKAR GOHIL
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| |
Collapse
|
20
|
Xu X, Xiao L, Xiao P, Yang S, Chen G, Liu F, Kanwar YS, Sun L. A glimpse of matrix metalloproteinases in diabetic nephropathy. Curr Med Chem 2015; 21:3244-60. [PMID: 25039784 DOI: 10.2174/0929867321666140716092052] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/06/2014] [Accepted: 07/11/2014] [Indexed: 12/14/2022]
Abstract
Matrix metalloproteinases (MMPs) are proteolytic enzymes belonging to the family of zinc-dependent endopeptidases that are capable of degrading almost all the proteinaceous components of the extracellular matrix (ECM). It is known that MMPs play a role in a number of renal diseases, such as, various forms of glomerulonephritis and tubular diseases, including some of the inherited kidney diseases. In this regard, ECM accumulation is considered to be a hallmark morphologic finding of diabetic nephropathy, which not only is related to the excessive synthesis of matrix proteins, but also to their decreased degradation by the MMPs. In recent years, increasing evidence suggest that there is a good correlation between the activity or expression of MMPs and progression of renal disease in patients with diabetic nephropathy and in various experimental animal models. In such a diabetic milieu, the expression of MMPs is modulated by high glucose, advanced glycation end products (AGEs), TGF-β, reactive oxygen species (ROS), transcription factors and some of the microRNAs. In this review, we focused on the structure and functions of MMPs, and their role in the pathogenesis of diabetic nephropathy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - L Sun
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan 415800, China..
| |
Collapse
|
21
|
Imbalance of the nerve growth factor and its precursor as a potential biomarker for diabetic retinopathy. BIOMED RESEARCH INTERNATIONAL 2015; 2015:571456. [PMID: 25853140 PMCID: PMC4380101 DOI: 10.1155/2015/571456] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 10/08/2014] [Accepted: 10/08/2014] [Indexed: 02/07/2023]
Abstract
Our previous studies have demonstrated that diabetes-induced oxidative stress alters homeostasis of retinal nerve growth factor (NGF) resulting in accumulation of its precursor, proNGF, at the expense of NGF which plays a critical role in preserving neuronal and retinal function. This imbalance coincided with retinal damage in experimental diabetes. Here we test the hypothesis that alteration of proNGF and NGF levels observed in retina and vitreous will be mirrored in serum of diabetic patients. Blood and vitreous samples were collected from patients (diabetic and nondiabetic) undergoing vitrectomy at Georgia Regents University under approved IRB. Levels of proNGF, NGF, and p75NTR shedding were detected using Western blot analysis. MMP-7 activity was also assayed. Diabetes-induced proNGF expression and impaired NGF expression were observed in vitreous and serum. Vitreous and sera from diabetic patients (n = 11) showed significant 40.8-fold and 3.6-fold increases, respectively, compared to nondiabetics (n = 9). In contrast, vitreous and sera from diabetic patients showed significant 44% and 64% reductions in NGF levels, respectively, compared to nondiabetics. ProNGF to NGF ratios showed significant correlation between vitreous and serum. Further characterization of diabetes-induced imbalance in the proNGF to NGF ratio will facilitate its utility as an early biomarker for diabetic complications.
Collapse
|
22
|
Ali S, Driscoll HE, Newton VL, Gardiner NJ. Matrix metalloproteinase-2 is downregulated in sciatic nerve by streptozotocin induced diabetes and/or treatment with minocycline: Implications for nerve regeneration. Exp Neurol 2014; 261:654-65. [PMID: 25158309 PMCID: PMC4199570 DOI: 10.1016/j.expneurol.2014.08.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 08/07/2014] [Accepted: 08/11/2014] [Indexed: 12/13/2022]
Abstract
Minocycline is an inhibitor of matrix metalloproteinases (MMPs) and has been shown to have analgesic effects. Whilst increased expression of MMPs is associated with neuropathic pain, MMPs also play crucial roles in Wallerian degeneration and nerve regeneration. In this study we examined the expression of MMP-2, MMP-9 and tissue inhibitor of metalloproteinase (TIMP)-1/-2 in the sciatic nerve of control and streptozotocin-induced diabetic rats treated with either vehicle or minocycline by quantitative PCR and gelatin zymography. We assessed the effects of minocycline on nerve conduction velocity and intraepidermal nerve fibre (IENF) deficits in diabetic neuropathy and investigated the effects of minocycline or MMP-2 on neurite outgrowth from primary cultures of dissociated adult rat sensory neurons. We show that MMP-2 is expressed constitutively in the sciatic nerve in vivo and treatment with minocycline or diabetes leads to downregulation of MMP-2 expression and activity. The functional consequence of this is IENF deficits in minocycline-treated nondiabetic rats and an unsupportive microenvironment for regeneration in diabetes. Minocycline reduces levels of MMP-2 mRNA and nerve growth factor-induced neurite outgrowth. Furthermore, in vivo minocycline treatment reduces preconditioning-induced in vitro neurite outgrowth following a sciatic nerve crush. In contrast, the addition of active MMP-2 facilitates neurite outgrowth in the absence of neurotrophic support and pre-treatment of diabetic sciatic nerve substrata with active MMP-2 promotes a permissive environment for neurite outgrowth. In conclusion we suggest that MMP-2 downregulation may contribute to the regenerative deficits in diabetes. Minocycline treatment also downregulates MMP-2 activity and is associated with inhibitory effects on sensory neurons. Thus, caution should be exhibited with its use as the balance between beneficial and detrimental outcomes may be critical in assessing the benefits of using minocycline to treat diabetic neuropathy. MMP-2, but not MMP-9, is constitutively expressed in the adult rat sciatic nerve. Levels of cleaved active MMP-2 are reduced in sciatic nerve of diabetic rats. Active MMP-2 potentiates neurite outgrowth from sensory neurons. Minocycline reduces levels of MMP-2 mRNA and impairs NGF-induced neurite growth. Minocycline did not prevent nerve dysfunction in experimental diabetic neuropathy.
Collapse
Affiliation(s)
- Sumia Ali
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Heather E Driscoll
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Victoria L Newton
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK
| | - Natalie J Gardiner
- Faculty of Life Sciences, University of Manchester, Oxford Road, Manchester M13 9PT, UK.
| |
Collapse
|
23
|
Abstract
An intact microcirculation is vital for diffusion of oxygen and nutrients and for removal of toxins of every organ and system in the human body. The functional and/or anatomical loss of microvessels is known as rarefaction, which can compromise the normal organ function and have been suggested as a possible starting point of several diseases. The purpose of this overview is to discuss the potential underlying mechanisms leading to renal microvascular rarefaction, and the potential consequences on renal function and on the progression of renal damage. Although the kidney is a special organ that receives much more blood than its metabolic needs, experimental and clinical evidence indicates that renal microvascular rarefaction is associated to prevalent cardiovascular diseases such as diabetes, hypertension, and atherosclerosis, either as cause or consequence. On the other hand, emerging experimental evidence using progenitor cells or angiogenic cytokines supports the feasibility of therapeutic interventions capable of modifying the progressive nature of microvascular rarefaction in the kidney. This overview will also attempt to discuss the potential renoprotective mechanisms of the therapeutic targeting of the renal microcirculation.
Collapse
Affiliation(s)
- Alejandro R Chade
- The Department of Physiology and Biophysics, Center for Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, Mississippi, USA.
| |
Collapse
|
24
|
Bondar' IA, Klimontov VV. The role of matrix metalloproteinases and their inhibitors in the development of renal fibrosis in the patients with diabetes mellitus. ACTA ACUST UNITED AC 2012. [DOI: 10.14341/probl201258139-44] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The accumulation of components of extracellular matrix in the glomerular and interstitial compartments of the kidneys is a characteristic feature of diabetic nephropathy. The leading role in the extracellular matrix catabolism is played by matrix metalloproteinases (MMP). The activity of these enzymes is regulated by a group of inhibitors including tissue metalloproteinase inhibitors, plasminogen activator inhibitor-1, etc. Both in vivo and in vitro studies have demonstrated that a reduction of MMP activities and/or an increase of expression of MMP tissue inhibitors in the glomerular and tubular cells result in the suppression of catabolism of the components of extracellular matrix under the hyperglycemic conditions. Both circulating and urinary MMP as well as their inhibitors are considered to be new potential markers of renal fibrosis associated with diabetes mellitus. It is concluded that the directed activation of MMP and neutralization of their inhibitors provide a promising tool for the treatment of diabetic nephropathy.
Collapse
|
25
|
CCN-2 is up-regulated by and mediates effects of matrix bound advanced glycated end-products in human renal mesangial cells. J Cell Commun Signal 2011; 5:193-200. [PMID: 21630131 DOI: 10.1007/s12079-011-0137-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2011] [Accepted: 05/02/2011] [Indexed: 01/28/2023] Open
Abstract
CCN-2, also known as connective tissue growth factor (CCN-2/CTGF) is a cysteine rich, extracellular matrix protein that acts as a pro-fibrotic cytokine in tissues in many diseases, including in diabetic nephropathy. We have published that soluble advanced glycation end products (AGEs), that are present in increased amounts in diabetes, induce CCN-2. However in vivo AGEs are known to be heavily tissue bound and whether matrix bound AGEs regulate CCN-2 has not been investigated. In this study we determined in human renal mesangial cells if CCN-2 is induced by matrix associated AGEs and if CCN-2 may then secondarily mediate effects of matrix AGEs on extracellular matrix expansion. Data generated show that CCN-2 mRNA and protein expression are induced by matrix bound AGEs, and in contrast, this was not the case for TGF-β1 mRNA regulation. Using CCN-2 adenoviral anti-sense it was found that CCN-2 mediated the up-regulation of fibronectin and the tissue inhibitor of matrix metalloproteinase, TIMP-1, that was caused by matrix bound AGEs. In conclusion, CCN-2 is induced by non-enzymatically glycated matrix and it mediates downstream fibronectin and TIMP-1 increases, thus through this mechanism potentially contributing to ECM accumulation in the renal glomerulus in diabetes.
Collapse
|
26
|
Ali TK, Al-Gayyar MMH, Matragoon S, Pillai BA, Abdelsaid MA, Nussbaum JJ, El-Remessy AB. Diabetes-induced peroxynitrite impairs the balance of pro-nerve growth factor and nerve growth factor, and causes neurovascular injury. Diabetologia 2011; 54:657-68. [PMID: 20957344 DOI: 10.1007/s00125-010-1935-1] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 09/08/2010] [Indexed: 12/16/2022]
Abstract
AIMS/HYPOTHESIS Diabetic retinopathy, the leading cause of blindness in working-age Americans, is characterised by reduced neurotrophic support and increased proinflammatory cytokines, resulting in neurotoxicity and vascular permeability. We sought to elucidate how oxidative stress impairs homeostasis of nerve growth factor (NGF) and its precursor, proform of NGF (proNGF), to cause neurovascular dysfunction in the eye of diabetic patients. METHODS Levels of NGF and proNGF were examined in samples from human patients, from retinal Müller glial cell line culture cells and from streptozotocin-induced diabetic animals treated with and without atorvastatin (10 mg/kg daily, per os) or 5,10,15,20-tetrakis (4-sulfonatophenyl)porphyrinato iron (III) chloride (FeTPPs) (15 mg/kg daily, i.p.) for 4 weeks. Neuronal death and vascular permeability were assessed by TUNEL and extravasation of BSA-fluorescein. RESULTS Diabetes-induced peroxynitrite formation impaired production and activity of matrix metalloproteinase-7 (MMP-7), which cleaves proNGF extracellularly, leading to accumulation of proNGF and reducing NGF in samples from diabetic retinopathy patients and experimental models. Treatment of diabetic animals with atorvastatin exerted similar protective effects that blocked peroxynitrite using FeTPPs, restoring activity of MMP-7 and hence the balance between proNGF and NGF. These effects were associated with preservation of blood-retinal barrier integrity, preventing neuronal cell death and blocking activation of RhoA and p38 mitogen-activated protein kinase (p38MAPK) in experimental and human samples. CONCLUSIONS/INTERPRETATION Oxidative stress plays an unrecognised role in causing accumulation of proNGF, which can activate a common pathway, RhoA/p38MAPK, to mediate neurovascular injury. Oral statin therapy shows promise for treatment of diabetic retinopathy.
Collapse
Affiliation(s)
- T K Ali
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA 30912, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Bao W, Min D, Twigg SM, Shackel NA, Warner FJ, Yue DK, McLennan SV. Monocyte CD147 is induced by advanced glycation end products and high glucose concentration: possible role in diabetic complications. Am J Physiol Cell Physiol 2010; 299:C1212-9. [PMID: 20810913 DOI: 10.1152/ajpcell.00228.2010] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
CD147 is a highly glycosylated transmembrane protein that is known to play a role in regulation of many protein families. It has the unique ability to maintain functional activity in both the membrane bound state and in the soluble form. CD147 is known to play a role in regulation of matrix metalloproteinase (MMP) expression, but whether its expression is affected by the diabetic milieu is not known, and its role in regulation of monocyte MMPs in this environment has not been investigated. Therefore, in this study we investigated the effect of advanced glycation end products (AGEs) and high glucose (HG; 25 mM), on monocyte CD147 expression. Culture of THP-1 monocytes in the presence of AGEs or HG significantly increased CD147 at the gene and protein level. THP-1 cell results were confirmed using freshly isolated monocytes from human volunteers. The effect of AGEs and HG on CD147 expression was also mimicked by addition of proinflammatory cytokines. Addition of AGEs or HG also increased expression of monocyte MMP-1 and MMP-9 but not MMP-2. This increase in MMPs was significantly attenuated by inhibition of CD147 using either a small interfering RNA or an anti-CD147 antibody. Inhibition of NF-κB or addition of antibodies to either TNF-α or the receptor for AGE (RAGE) each significantly prevented in a dose-dependent manner the induction of CD147 gene and protein by AGE and also decreased MMP-1 and MMP-9. This novel result shows that AGEs can induce monocyte CD147 expression, an effect mediated by inflammatory pathways and RAGE. Because MMPs play a role in monocyte migration, inhibition of their regulator CD147 may assist in the prevention of diabetic complications, particularly those where monocyte infiltration is an early initiating event.
Collapse
Affiliation(s)
- W Bao
- Discipline of Medicine and Bosch Institute, University of Sydney, Sydney, New South Wales, Australia
| | | | | | | | | | | | | |
Collapse
|
28
|
Chiao YA, Zamilpa R, Lopez EF, Dai Q, Escobar GP, Hakala K, Weintraub ST, Lindsey ML. In vivo matrix metalloproteinase-7 substrates identified in the left ventricle post-myocardial infarction using proteomics. J Proteome Res 2010; 9:2649-57. [PMID: 20232908 DOI: 10.1021/pr100147r] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Matrix metalloproteinase-7 (MMP-7) deletion has been shown to improve survival after myocardial infarction (MI). MMP-7 has a large array of in vitro substrates, but in vivo substrates for MMP-7 following MI have not been fully identified. Accordingly, we evaluated the infarct regions of wild-type (WT; n = 12) and MMP-7 null (null; n = 10) mice using a proteomic strategy. Seven days post-MI, infarct regions of the left ventricles were excised, homogenized, and protein extracts were analyzed by two-dimensional gel electrophoresis and mass spectrometry. Of 13 spots that showed intensity differences between WT and null, the intensities of eight spots were higher and those of five spots were lower in the null group (p < 0.05). Fibronectin and tenascin-C, known in vitro substrates of MMP-7, were identified in spots that showed lower intensity in the null. Immunoblotting and in vitro cleavage assays confirmed reduced fibronectin and tenascin-C fragment generation in the null, and this effect was restored by exogenous administration of MMP-7. Lower levels of full-length peroxiredoxin-1 and -2 and higher levels of the full-length peroxiredoxin-3 were detected in the null group, suggesting MMP-7 deletion may also indirectly regulate protein levels through nonenzymatic mechanisms. In conclusion, this is the first study to identify fibronectin and tenascin-C as in vivo MMP-7 substrates in the infarcted left ventricle using a proteomic approach.
Collapse
Affiliation(s)
- Ying Ann Chiao
- Department of Medicine, Division of Cardiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Ban CR, Twigg SM, Franjic B, Brooks BA, Celermajer D, Yue DK, McLennan SV. Serum MMP-7 is increased in diabetic renal disease and diabetic diastolic dysfunction. Diabetes Res Clin Pract 2010; 87:335-41. [PMID: 20096949 DOI: 10.1016/j.diabres.2010.01.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Revised: 12/11/2009] [Accepted: 01/04/2010] [Indexed: 01/01/2023]
Abstract
Circulating matrix metalloproteinase (MMP) levels may correlate with diabetic complications. Whether they are changed in early diabetic cardiomyopathy is not known and was examined in this study. TIMP-1 and collagen degradation products were also measured. Results from subjects with and without diastolic dysfunction were compared with those obtained for patients with varying stages of diabetic renal disease. Patients with type 2 diabetes with or without diastolic dysfunction with varying degrees of renal disease were recruited for this study. Age-matched non-diabetic subjects served as controls. MMPs (-1, -3 and -7) and TIMP-1 were measured by ELISA, MMP-2 and -9 by zymography and collagen degradation products by radioimmunoassay. Differences in the pattern of MMPs/TIMPs and collagen degradation products were observed. The most consistent change was in totalMMP-7, which was increased in those with diastolic dysfunction and those with macroalbuminuria. MMP-7 correlated with cardiac function (p<0.05 vs control, in those with diastolic dysfunction), and renal filtration function (p<0.05 vs control). In summary, we have identified novel relationships between serum MMP-7 and diabetic complications specifically in renal disease and in diastolic dysfunction. How increased circulating MMP-7 is associated with these diabetic microvascular complications and the significance of these findings will require prospective studies.
Collapse
Affiliation(s)
- C R Ban
- Alesd Hospital, Bihor County, Romania
| | | | | | | | | | | | | |
Collapse
|
30
|
Daroux M, Prévost G, Maillard-Lefebvre H, Gaxatte C, D’Agati V, Schmidt A, Boulanger É. Advanced glycation end-products: Implications for diabetic and non-diabetic nephropathies. DIABETES & METABOLISM 2010; 36:1-10. [DOI: 10.1016/j.diabet.2009.06.005] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2009] [Revised: 06/21/2009] [Accepted: 06/23/2009] [Indexed: 01/28/2023]
|
31
|
Aggarwal HK, Jain D, Talapatra P, Yadav RK, Gupta T, Kathuria KL. Evaluation of role of doxycycline (a matrix metalloproteinase inhibitor) on renal functions in patients of diabetic nephropathy. Ren Fail 2010; 32:941-946. [PMID: 20722561 DOI: 10.3109/0886022x.2010.502606] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
This study was conducted to see the effect of doxycycline on renal functions, especially proteinuria, in patients of diabetic nephropathy (DN). The study included 40 clinically proven adult patients of DN. All patients were on stable doses of angiotensin-converting enzyme inhibitors (ACEIs) and or angiotensin receptor blockers (ARBs) for 2 months before the study. The patients were divided into two groups of 20 patients each. Group A patients were maintained on stable dose of ACEIs and/or ARBs, whereas Group B patients received doxycycline (100 mg/day) for a period of 3 months in addition to ACEIs and or ARBs. Adequate glycemic control was achieved with insulin or oral hypoglycemic agents in all the patients. Renal parameters were assessed at the beginning of the study, at 1, 3, and 6 months (after a washout period of 3 months). All renal parameters remained unaltered during the study in both groups. However, proteinuria showed improvement in Group B (doxcycycline group).The mean basal value of proteinuria was 1.74 + 1.70 for Group A and 2.17 + 2.95 for Group B. At the end of 3 months, proteinuria was 1.22 + 2.11 in Group B whereas it was 1.50 + 1.50 in Group A (p < 0.05). However, the decrease in proteinuria at 6 months in the two groups did not show any statistically significant difference. No significant side effects of doxycycline were observed. The study showed that doxycycline was effective in reducing proteinuria in patients of DN when used for the short duration of 3 months.
Collapse
Affiliation(s)
- Hari Krishan Aggarwal
- Department of Medicine, Division of Nephrology, Pt. B.D. Sharma University of Health Sciences, Rohtak, Haryana, India.
| | | | | | | | | | | |
Collapse
|
32
|
Xiao J, Lv Y, Lin S, Jin L, Zhang Y, Wang X, Ma J, Hu K, Feng W, Cai L, Li X, Tan Y. Cardiac protection by basic fibroblast growth factor from ischemia/reperfusion-induced injury in diabetic rats. Biol Pharm Bull 2010; 33:444-449. [PMID: 20190407 DOI: 10.1248/bpb.33.444] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2024]
Abstract
Diabetes impairs the expression and function of endogenous growth factors, leading to increased cardiovascular events in diabetic patients. Supplementation of fibroblast growth factors (FGFs) protected the heart from ischemia/reperfusion (I/R)-induced injury in animal models. However, it has not yet been tested in diabetic heart. The present study was thus to clarify whether basic fibroblast growth factor (bFGF) could protect the heart from I/R-induced damage under diabetic conditions using a rat model. Male Sprague Dawley rats were used to induce diabetes by intraperitoneal injection of streptozotocin. Eight weeks later, I/R injury was generated in diabetic rats and age-matched non-diabetic rats. All I/R rats were administrated bFGF or saline through intramyocardial injection. Seven days after I/R, cardiac infarction, structural changes, cell death and blood vessel density, serum malondialdehyde (MDA) and cardiac enzyme lactate dehydrogenase (LDH) were examined. We found that I/R induced significant increases in the cardiac infarction, blood MDA contents and LDH activities, and the expression of caspase-3. Treatment of I/R rats with bFGF simultaneously with reperfusion significantly attenuated I/R-induced pathological changes, along with a significant increase in the cardiac blood vessel density in both diabetic and non-diabetic rates. The protective effects of bFGF on I/R-induced cardiac injury in diabetic group are less than those in non-diabetic group. The results indicated that bFGF provide a protection of the heart against I/R-induced oxidative damage, cell death and infarction under diabetic conditions.
Collapse
Affiliation(s)
- Jian Xiao
- Chinese-American Research Institute for Diabetic Complications, Wenzhou Medical College, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Roy S, Trudeau K, Roy S, Behl Y, Dhar S, Chronopoulos A. New Insights into Hyperglycemia-induced Molecular Changes in Microvascular Cells. J Dent Res 2009; 89:116-27. [DOI: 10.1177/0022034509355765] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Hyperglycemia is the most prevalent characteristic of diabetes and plays a central role in mediating adverse effects on vascular cells during the progression of diabetic vascular complications. In diabetic microangiopathy, hyperglycemia induces biochemical and molecular changes in microvascular cells that ultimately progress to retinal, renal, and neural complications and extends to other complications, including advanced periodontal disease. In this review, we describe changes involving basement membrane thickening, tissue remodeling, gap junctions, inflammation, cytokines, and transcription factors, and their effects on the pathogenesis of diabetic microvascular complications. The majority of the changes described relate to retinal microangiopathy, since ultrastructural, structural, and biochemical alterations have been well-characterized in this tissue.
Collapse
Affiliation(s)
- S. Roy
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston University, 650 Albany Street, Boston, MA 02118, USA
| | - K. Trudeau
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston University, 650 Albany Street, Boston, MA 02118, USA
| | - S. Roy
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston University, 650 Albany Street, Boston, MA 02118, USA
| | - Y. Behl
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston University, 650 Albany Street, Boston, MA 02118, USA
| | - S. Dhar
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston University, 650 Albany Street, Boston, MA 02118, USA
| | - A. Chronopoulos
- Departments of Medicine and Ophthalmology, Boston University School of Medicine, and
- Department of Periodontology and Oral Biology, Boston University School of Dental Medicine, Boston University, 650 Albany Street, Boston, MA 02118, USA
| |
Collapse
|
34
|
Ribeiro DL, Taboga SR, Góes RM. Diabetes induces stromal remodelling and increase in chondroitin sulphate proteoglycans of the rat ventral prostate. Int J Exp Pathol 2009; 90:400-11. [PMID: 19659898 DOI: 10.1111/j.1365-2613.2009.00657.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Extracellular matrix (ECM) remodelling is an important process involved in prostate cancer progression. Alterations in ECM caused by diabetes in different tissues such as kidney is well described; however, it is poorly investigated in prostate. The aim of this study was to evaluate changes in ECM of rat prostate showing gland atrophy caused by diabetes and their implications in development of malignant lesions. Diabetes was induced in Wistar rats using alloxan (45 mg/kg bw). After 90 days of diabetes onset, animals were killed and ventral prostate was removed and prepared for light microscopy following immunoreaction for fibronectin, chondroitin sulphate and Picrossirius staining for collagen fibres. Proteoglycans (PG) were identified at transmission electron microscopy after fixation with Cuprolinic Blue. Diabetes led to a thickening of 25% in the acinar basement membrane accompanied by increase and disorganization of its proteoglycans (P1). Three additional populations of prostatic stromal PGs were identified: collagen fibril linked (P2) and interstitial (P3) and (P4) PGs. Diabetes increased P3 and mainly P4 which had higher dimension and accumulated around the smooth muscle cells. In addition, an increase in chondrotin sulphate (33%, mainly in sites where P4 were noted) and collagen (44%) was noted in diabetic rats, whereas fibronectin did not change. Atrophic changes observed in rat ventral prostate after diabetes are accompanied by stromal remodelation related to increase in collagen and chondroitin sulphate proteoglycans. Thus, diabetes can promote a stromal microenvironment rich in elements that could favour cell migration, proliferation and pathological process.
Collapse
Affiliation(s)
- Daniele Lisboa Ribeiro
- Department of Cell Biology, Institute of Biology, State University of Campinas-Unicamp, Campinas, São Paulo, Brazil
| | | | | |
Collapse
|
35
|
Chen YD, Xu X, Xia X, Wu H, Liu K, Zheng Z, Zhu D. MMP9 Is Involved in Glycation End-Products Induced Increase of Retinal Vascular Permeability in Rats and the Therapeutic Effect of Minocycline. Curr Eye Res 2009; 33:977-83. [DOI: 10.1080/02713680802450984] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
36
|
|
37
|
Thrailkill KM, Clay Bunn R, Fowlkes JL. Matrix metalloproteinases: their potential role in the pathogenesis of diabetic nephropathy. Endocrine 2009; 35:1-10. [PMID: 18972226 PMCID: PMC2629499 DOI: 10.1007/s12020-008-9114-6] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Revised: 07/21/2008] [Accepted: 09/03/2008] [Indexed: 12/11/2022]
Abstract
Matrix metalloproteinases (MMPs), a family of proteinases including collagenases, gelatinases, stromelysins, matrilysins, and membrane-type MMPs, affect the breakdown and turnover of extracellular matrix (ECM). Moreover, they are major physiologic determinants of ECM degradation and turnover in the glomerulus. Renal hypertrophy and abnormal ECM deposition are hallmarks of diabetic nephropathy (DN), suggesting that altered MMP expression or activation contributes to renal injury in DN. Herein, we review and summarize recent information supporting a role for MMPs in the pathogenesis of DN. Specifically, studies describing dysregulated activity of MMPs and/or their tissue inhibitors in various experimental models of diabetes, including animal models of type 1 or type 2 diabetes, clinical investigations of human type 1 or type 2 diabetes, and kidney cell culture studies are reviewed.
Collapse
Affiliation(s)
- Kathryn M Thrailkill
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72202, USA.
| | | | | |
Collapse
|
38
|
Tveita A, Rekvig OP, Zykova SN. Glomerular matrix metalloproteinases and their regulators in the pathogenesis of lupus nephritis. Arthritis Res Ther 2008; 10:229. [PMID: 19090960 PMCID: PMC2656222 DOI: 10.1186/ar2532] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Lupus nephritis is a major contributor to morbidity and mortality in systemic lupus erythematosus, but little is known about the pathogenic processes that underlie the progressive decay in renal function. A common finding in lupus nephritis is thickening of glomerular basement membranes associated with immune complex deposition. It has been speculated that alterations in the synthesis or degradation of membrane components might contribute to such changes, and thereby to initiation and progression of nephritis through facilitation of immune complex deposition. Matrix metalloproteinases (MMPs) are enzymes that are intimately involved in the turnover of major glomerular basement membrane constituents, including collagen IV and laminins. Alterations in the expression and activity of MMPs have been described in a number of renal diseases, suggesting their relevance to the pathogenesis of various glomerulopathies. The same is true for their natural inhibitors, the tissue inhibitor of metalloproteinase family. Recent data from our group have identified an increase in proteolytic activity within the glomerulus coinciding with the development of proteinuria in the mouse model of systemic lupus erythematosus. (NXB x NZW)F1 Here we review current understanding of MMP/tissue inhibitor of metalloproteinase function within the kidney, and discuss their possible involvement in the development and progression of lupus nephritis.
Collapse
Affiliation(s)
- Anders Tveita
- Department of Biochemistry, Medical Faculty, Institute of Medical Biology, University of Tromsø, Tromsø, Norway.
| | | | | |
Collapse
|
39
|
van Golde JM, Ruiter MS, Schaper NC, Vöö S, Waltenberger J, Backes WH, Post MJ, Huijberts MS. Impaired collateral recruitment and outward remodeling in experimental diabetes. Diabetes 2008; 57:2818-23. [PMID: 18633114 PMCID: PMC2551694 DOI: 10.2337/db08-0229] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2008] [Accepted: 07/01/2008] [Indexed: 12/14/2022]
Abstract
OBJECTIVE In this study, the effect of chronic hyperglycemia on acute ligation-induced collateral vasodilation, on monocyte chemotaxis, and on structural outward remodeling of collaterals was investigated. RESEARCH DESIGN AND METHODS Femoral artery ligation was performed 8 weeks after alloxan or saline treatment in New Zealand White rabbits. Angiography was performed directly, 1 and 3 weeks after ligation. These angiographic recordings were used to quantify number of collaterals, lumen, and blood volume index. Reactive hyperemia response was tested by intramuscular laser Doppler measurements. Subsequently, blood was sampled from the aorta for monocyte chemotaxis. RESULTS Ligation resulted in markedly lower acute collateral vasodilation in diabetic compared with control rabbits. Also, hyperemic vasodilatory response to local ischemia was impaired in diabetic rabbits. This difference persisted at 1 and 3 weeks after ligation, with a lower number of visible collaterals. In addition, the collateral lumen was markedly lower in diabetic rabbits after the maturation phase. Likewise, a reduced blood volume index in the region of growing collaterals was observed in diabetic animals. The monocyte migration toward vascular endothelial growth factor-A and monocyte chemotactic protein-1 was strongly reduced in diabetic rabbits. CONCLUSIONS This study demonstrates that chronic hyperglycemia negatively affects the different phases of arteriogenesis: 1) impaired shear induced vasodilatation; 2) impaired outward collateral growth, reflected in the number of collaterals and blood volume index; and 3) inhibition of monocyte chemotaxis. Impairments were most evident in the acute phase of arteriogenesis. Therapies aimed at restoring acute collateral recruitment, such as vasodilators, may be of interest to improve collateral function in diabetes.
Collapse
Affiliation(s)
- Jolanda M van Golde
- Department of Internal Medicine, Division of Endocrinology, Maastricht University Medical Centre and Cardiovascular Research Institute Maastricht, Maastricht, the Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Cohen CD, Lindenmeyer MT, Eichinger F, Hahn A, Seifert M, Moll AG, Schmid H, Kiss E, Gröne E, Gröne HJ, Kretzler M, Werner T, Nelson PJ. Improved elucidation of biological processes linked to diabetic nephropathy by single probe-based microarray data analysis. PLoS One 2008; 3:e2937. [PMID: 18698414 PMCID: PMC2493035 DOI: 10.1371/journal.pone.0002937] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2008] [Accepted: 07/09/2008] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a complex and chronic metabolic disease that evolves into a progressive fibrosing renal disorder. Effective transcriptomic profiling of slowly evolving disease processes such as DN can be problematic. The changes that occur are often subtle and can escape detection by conventional oligonucleotide DNA array analyses. METHODOLOGY/PRINCIPAL FINDINGS We examined microdissected human renal tissue with or without DN using Affymetrix oligonucleotide microarrays (HG-U133A) by standard Robust Multi-array Analysis (RMA). Subsequent gene ontology analysis by Database for Annotation, Visualization and Integrated Discovery (DAVID) showed limited detection of biological processes previously identified as central mechanisms in the development of DN (e.g. inflammation and angiogenesis). This apparent lack of sensitivity may be associated with the gene-oriented averaging of oligonucleotide probe signals, as this includes signals from cross-hybridizing probes and gene annotation that is based on out of date genomic data. We then examined the same CEL file data using a different methodology to determine how well it could correlate transcriptomic data with observed biology. ChipInspector (CI) is based on single probe analysis and de novo gene annotation that bypasses probe set definitions. Both methods, RMA and CI, used at default settings yielded comparable numbers of differentially regulated genes. However, when verified by RT-PCR, the single probe based analysis demonstrated reduced background noise with enhanced sensitivity and fewer false positives. CONCLUSIONS/SIGNIFICANCE Using a single probe based analysis approach with de novo gene annotation allowed an improved representation of the biological processes linked to the development and progression of DN. The improved analysis was exemplified by the detection of Wnt signaling pathway activation in DN, a process not previously reported to be involved in this disease.
Collapse
Affiliation(s)
- Clemens D. Cohen
- Nephrology Clinic and Institute of Physiology with Center of Integrative Human Physiology, University Hospital and University, Zurich, Switzerland
- Clinical Biochemistry Group, Medical Policlinic, University of Munich, Munich, Germany
| | - Maja T. Lindenmeyer
- Nephrology Clinic and Institute of Physiology with Center of Integrative Human Physiology, University Hospital and University, Zurich, Switzerland
- Clinical Biochemistry Group, Medical Policlinic, University of Munich, Munich, Germany
| | - Felix Eichinger
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | | | | | - Anton G. Moll
- Nephrology Clinic and Institute of Physiology with Center of Integrative Human Physiology, University Hospital and University, Zurich, Switzerland
- Clinical Biochemistry Group, Medical Policlinic, University of Munich, Munich, Germany
| | - Holger Schmid
- Clinical Biochemistry Group, Medical Policlinic, University of Munich, Munich, Germany
| | - Eva Kiss
- Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Elisabeth Gröne
- Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Hermann-Josef Gröne
- Cellular and Molecular Pathology, German Cancer Research Center, Heidelberg, Germany
| | - Matthias Kretzler
- Division of Nephrology, Department of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
| | | | - Peter J. Nelson
- Clinical Biochemistry Group, Medical Policlinic, University of Munich, Munich, Germany
- * E-mail:
| |
Collapse
|
41
|
Samuel CS, Hewitson TD, Zhang Y, Kelly DJ. Relaxin ameliorates fibrosis in experimental diabetic cardiomyopathy. Endocrinology 2008; 149:3286-93. [PMID: 18388190 DOI: 10.1210/en.2008-0250] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Fibrosis (extracellular matrix accumulation) is the final end point in diabetic cardiomyopathy. The current study evaluated the therapeutic effects of the antifibrotic hormone relaxin (RLX) in streptozotocin-treated transgenic mRen-2 rats, which undergo pathological and functional features similar to human diabetes. Twelve-week-old hyperglycemic mRen-2 rats, normoglycemic control rats, and animals treated with recombinant human gene-2 (H2) RLX from wk 10-12 were assessed for various measures of left ventricular (LV) fibrosis, hemodynamics, and function, while the mechanism of RLX's actions was also determined. Hyperglycemic mRen-2 rats had increased LV collagen concentration (fibrosis) and gelatinase activity (all P < 0.05 vs. controls) but equivalent levels of interstitial collagenase and tissue inhibitor of metalloproteinase-1 to that measured in control rats. The increased LV fibrosis associated with diabetic animals led to significant alterations in the E/A wave ratio and E-wave deceleration time (both P < 0.05 vs. controls) in the absence of blood pressure changes, reflective of myocardial stiffness and LV diastolic dysfunction. H2-RLX treatment of diabetic rats led to significant decreases in interstitial and total LV collagen deposition (both P < 0.05 vs. diabetic group), resulting in decreased myocardial stiffness and improved LV diastolic function, without affecting nondiabetic animals. The protective effects of H2-RLX in diabetic rats were associated with a reduction in mesenchymal cell differentiation and tissue inhibitor of metalloproteinase-1 expression in addition to a promotion of extracellular matrix-degrading matrix metalloproteinase-13 (all P < 0.05 vs. diabetic group) but were independent of blood pressure regulation. These findings demonstrate that RLX is an antifibrotic with rapid-occurring efficacy and may represent a novel therapy for the treatment of diabetes.
Collapse
Affiliation(s)
- Chrishan S Samuel
- Howard Florey Institute and Department of Biochemistry and Molecular Biology, University of Melbourne, Victoria 3010, Australia.
| | | | | | | |
Collapse
|
42
|
Silverstein DM, Craver RD. Mesangial hypercellularity in children: presenting features and outcomes. Pediatr Nephrol 2008; 23:921-8. [PMID: 18324424 DOI: 10.1007/s00467-008-0750-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2007] [Revised: 12/03/2007] [Accepted: 12/07/2007] [Indexed: 10/22/2022]
Abstract
Mesangial hypercellularity (MH), in the absence of sclerosis or immune deposition, was a common finding on renal biopsy in our center. We studied 66 children with predominant MH. Among all patients older than 2.7 years, blood pressure (BP) percentile and glomerular filtration rate (GFR) remained stable. Serum albumin (Alb) trended higher (3.0+/-0.2 start vs. 3.4+/-0.2 end g/dl, p=0.06) and urine protein/creatinine lower (4.2+/-0.9 start vs. 2.3+/-0.9 end mg/mg, p=0.18) at the end of the study period. The proportion with stage 1 CKD remained constant: 94% start vs. 92% end. At end, Alb was lower in patients referred for nephrotic syndrome (NS): 4.4+/-0.3 hematuria vs. 4.2+/-0.2 proteinuria vs. 2.8+/-0.3 NS g/dl, p<0.05 vs. both. Alb was lower (p=0.03) and urine protein/creatinine trended higher in patients with diffuse foot-process fusion (FPF). Twenty-five percent of patients with focal FPF developed NS, all had relapses, and 63% were steroid sensitive (SS). All but one with diffuse FPF presented with NS; 86% had relapses (mean 1 year) and 63% were SS. GFR trended higher at the end in those with matrix thickening (mat) (119.6+/-4.7 no mat vs. 129.1+/-2.6 mat ml/min per 1.73 m2, p=0.1). Those without mat were less SS (59% no mat vs. 80% mat) and were more likely to require alkylating agents (Alk) for NS. Among those with positive immunofluorescence (IF), 82% had immunoglobulin M (IgM) alone; those with positive IF were more SS and needed Alk for NS. MH predicts a favorable prognosis. FPF predicts NS and multiple relapses.
Collapse
Affiliation(s)
- Douglas M Silverstein
- Department of Pediatrics, Louisiana State University Health Sciences Center and Children's Hospital New Orleans, New Orleans, LA 70118, USA.
| | | |
Collapse
|