1
|
Srivastava A, Maienschein-Cline M, Amdur RL, Susztak K, Kopp JB, Cao H, Shankaranarayanan D, Abdalla Y, Pabalan A, Gombert M, Raj D. Plasma Transcriptome Profile Associated with Chronic Kidney Disease Progression. Am J Nephrol 2024; 56:296-308. [PMID: 39694024 DOI: 10.1159/000542707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 11/14/2024] [Indexed: 12/20/2024]
Abstract
INTRODUCTION Understanding the molecular signals associated with the progression of kidney disease is vital for risk stratification and targeted treatment. Recent advances in RNA-sequencing technique have enabled us to characterize extracellular transcriptome profiles for precision diagnostics. METHOD We evaluated the plasma mRNA profile of participants exhibiting slow (n = 119) and fast (n = 119) decline in estimated glomerular filtration rate (eGFR) among the Chronic Renal Insufficiency Cohort (CRIC) in a nested case control study. The two groups were matched for age, sex, race, baseline eGFR, proteinuria, and diabetes status. The next-generation sequencing data were analyzed using edgeR to identify differentially expressed genes (DEGs) and ingenuity pathway analysis was done to identify the associated pathways. We also compared the top plasma DEGs with gene expression in microdissected human chronic kidney disease (CKD) kidney. RESULTS We identified fragments from ∼28,000 annotated genes, of which 783 transcripts exhibited differential expression between slow and fast CKD progressors. Among 629 protein coding genes, 469 were overexpressed in slow progressors, while 157 showed increased expression in fast progressors. Expression of GLI2, CUX1, NOTCH1, and LRP1 transcripts were amplified in slow progressors. Pathway analysis linked these DEG to WNT/β-catenin signaling, IL-12 signaling and production in macrophages, Netrin-1 Signaling and Epithelial-Mesenchymal Transition pathways. Many of the plasma DEGs were also upregulated in microdissected human CKD kidney. CONCLUSION Warranting further validation, circulating levels of aberrantly expressed transcripts hold potential to be used as biomarkers for fast CKD progression.
Collapse
Affiliation(s)
- Anvesha Srivastava
- Division of Renal Diseases and Hypertension, George Washington University, Washington, DC, USA
| | - Mark Maienschein-Cline
- Research Informatics Core, University of Illinois at Chicago Research Resources Center, Chicago, Illinois, USA
| | - Richard L Amdur
- Feinstein Institute for Medical Research, Northwell Health, Manhasset, New York, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Institute of Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Jeffrey Burnett Kopp
- Kidney Disease Section, Kidney Diseases Branch, National Institute of Diabetes and Digestive and Kidney disease, National Institutes of Health, Bethesda, Maryland, USA
| | - Haiming Cao
- Cardiovascular Branch, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Divya Shankaranarayanan
- Division of Renal Diseases and Hypertension, George Washington University, Washington, DC, USA
| | - Yoosif Abdalla
- Division of Renal Diseases and Hypertension, George Washington University, Washington, DC, USA
| | - Ana Pabalan
- Division of Renal Diseases and Hypertension, George Washington University, Washington, DC, USA
| | | | - Dominic Raj
- Division of Renal Diseases and Hypertension, George Washington University, Washington, DC, USA
| |
Collapse
|
2
|
Singh B, Cui K, Eisa-Beygi S, Zhu B, Cowan DB, Shi J, Wang DZ, Liu Z, Bischoff J, Chen H. Elucidating the crosstalk between endothelial-to-mesenchymal transition (EndoMT) and endothelial autophagy in the pathogenesis of atherosclerosis. Vascul Pharmacol 2024; 155:107368. [PMID: 38548093 PMCID: PMC11303600 DOI: 10.1016/j.vph.2024.107368] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/07/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024]
Abstract
Atherosclerosis, a chronic systemic inflammatory condition, is implicated in most cardiovascular ischemic events. The pathophysiology of atherosclerosis involves various cell types and associated processes, including endothelial cell activation, monocyte recruitment, smooth muscle cell migration, involvement of macrophages and foam cells, and instability of the extracellular matrix. The process of endothelial-to-mesenchymal transition (EndoMT) has recently emerged as a pivotal process in mediating vascular inflammation associated with atherosclerosis. This transition occurs gradually, with a significant portion of endothelial cells adopting an intermediate state, characterized by a partial loss of endothelial-specific gene expression and the acquisition of "mesenchymal" traits. Consequently, this shift disrupts endothelial cell junctions, increases vascular permeability, and exacerbates inflammation, creating a self-perpetuating cycle that drives atherosclerotic progression. While endothelial cell dysfunction initiates the development of atherosclerosis, autophagy, a cellular catabolic process designed to safeguard cells by recycling intracellular molecules, is believed to exert a significant role in plaque development. Identifying the pathological mechanisms and molecular mediators of EndoMT underpinning endothelial autophagy, may be of clinical relevance. Here, we offer new insights into the underlying biology of atherosclerosis and present potential molecular mechanisms of atherosclerotic resistance and highlight potential therapeutic targets.
Collapse
Affiliation(s)
- Bandana Singh
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Kui Cui
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Shahram Eisa-Beygi
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Bo Zhu
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Douglas B Cowan
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Jinjun Shi
- Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Da-Zhi Wang
- Center for Regenerative Medicine, University of South Florida Health Heart Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Zhenguo Liu
- Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, USA
| | - Joyce Bischoff
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Hong Chen
- Vascular Biology Program, Department of Surgery, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
3
|
Zhu J, Li Q, Sun Y, Zhang S, Pan R, Xie Y, Chen J, Shi L, Chen Y, Sun Z, Zhang L. Insulin-Like Growth Factor 1 Receptor Deficiency Alleviates Angiotensin II-Induced Cardiac Fibrosis Through the Protein Kinase B/Extracellular Signal-Regulated Kinase/Nuclear Factor-κB Pathway. J Am Heart Assoc 2023; 12:e029631. [PMID: 37721135 PMCID: PMC10547288 DOI: 10.1161/jaha.123.029631] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 08/15/2023] [Indexed: 09/19/2023]
Abstract
Background The renin-angiotensin system plays a crucial role in the development of heart failure, and Ang II (angiotensin II) acts as the critical effector of the renin-angiotensin system in regulating cardiac fibrosis. However, the mechanisms of cardiac fibrosis are complex and still not fully understood. IGF1R (insulin-like growth factor 1 receptor) has multiple functions in maintaining cardiovascular homeostasis, and low-dose IGF1 treatment is effective in relieving Ang II-induced cardiac fibrosis. Here, we aimed to investigate the molecular mechanism of IGF1R in Ang II-induced cardiac fibrosis. Methods and Results Using primary mouse cardiac microvascular endothelial cells and fibroblasts, in vitro experiments were performed. Using C57BL/6J mice and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 (Cas9)-mediated IGF1R heterozygous knockout (Igf1r+/-) mice, cardiac fibrosis mouse models were induced by Ang II for 2 weeks. The expression of IGF1R was examined by quantitative reverse transcription polymerase chain reaction, immunohistochemistry, and Western blot. Mice heart histologic changes were evaluated using Masson and picro sirius red staining. Fibrotic markers and signal molecules indicating the function of the Akt (protein kinase B)/ERK (extracellular signal-regulated kinase)/nuclear factor-κB pathway were detected using quantitative reverse transcription polymerase chain reaction and Western blot. RNA sequencing was used to explore IGF1R-mediated target genes in the hearts of mice, and the association of IGF1R and G-protein-coupled receptor kinase 5 was identified by coimmunoprecipitation. More important, blocking IGF1R signaling significantly suppressed endothelial-mesenchymal transition in primary mouse cardiac microvascular endothelial cells and mice in response to transforming growth factor-β1 or Ang II, respectively. Deficiency or inhibition of IGF1R signaling remarkably attenuated Ang II-induced cardiac fibrosis in primary mouse cardiac fibroblasts and mice. We further observed that the patients with heart failure exhibited higher blood levels of IGF1 and IGF1R than healthy individuals. Moreover, Ang II treatment significantly increased cardiac IGF1R in wild type mice but led to a slight downregulation in Igf1r+/- mice. Interestingly, IGF1R deficiency significantly alleviated cardiac fibrosis in Ang II-treated mice. Mechanistically, the phosphorylation level of Akt and ERK was upregulated in Ang II-treated mice, whereas blocking IGF1R signaling in mice inhibited these changes of Akt and ERK phosphorylation. Concurrently, phosphorylated p65 of nuclear factor-κB exhibited similar alterations in the corresponding group of mice. Intriguingly, IGF1R directly interacted with G-protein-coupled receptor kinase 5, and this association decreased ≈50% in Igf1r+/- mice. In addition, Grk5 deletion downregulated expression of the Akt/ERK/nuclear factor-κB signaling pathway in primary mouse cardiac fibroblasts. Conclusions IGF1R signaling deficiency alleviates Ang II-induced cardiac fibrosis, at least partially through inhibiting endothelial-mesenchymal transition via the Akt/ERK/nuclear factor-κB pathway. Interestingly, G-protein-coupled receptor kinase 5 associates with IGF1R signaling directly, and it concurrently acts as an IGF1R downstream effector. This study suggests the promising potential of IGF1R as a therapeutic target for cardiac fibrosis.
Collapse
Affiliation(s)
- Jiafeng Zhu
- Department of NursingWeifang Medical UniversityWeifangChina
| | - Qian Li
- Department of NursingWeifang Medical UniversityWeifangChina
| | - Yan Sun
- Department of StomatologyWeifang Medical UniversityWeifangChina
| | - Shiyu Zhang
- Department of NursingWeifang Medical UniversityWeifangChina
| | - Ruiyan Pan
- Department of PharmacologyWeifang Medical UniversityWeifangChina
| | - Yanguang Xie
- Department of NursingWeifang Medical UniversityWeifangChina
| | - Jinyan Chen
- Department of Clinical MedicineWeifang Medical UniversityWeifangChina
| | - Lihong Shi
- Department of Rehabilitation MedicineWeifang Medical UniversityWeifangChina
| | - Yanbo Chen
- Department of Cardiology, The First Affiliated HospitalWeifang Medical UniversityWeifangChina
| | - Zhipeng Sun
- Department of PharmacologyWeifang Medical UniversityWeifangChina
| | - Lane Zhang
- Department of NursingWeifang Medical UniversityWeifangChina
| |
Collapse
|
4
|
Abstract
The endothelium is one of the largest organ systems in the body, and data continue to emerge regarding the importance of endothelial cell (EC) dysfunction in vascular aging and a range of cardiovascular diseases (CVDs). Over the last two decades and as a process intimately related to EC dysfunction, an increasing number of studies have also implicated endothelial to mesenchymal transition (EndMT) as a potentially disease-causal pathobiologic process that is involved in a multitude of differing CVDs. However, EndMT is also involved in physiologic processes (e.g., cardiac development), and transient EndMT may contribute to vascular regeneration in certain contexts. Given that EndMT involves a major alteration in the EC-specific molecular program, and that it potentially contributes to CVD pathobiology, the clinical translation opportunities are significant, but further molecular and translational research is needed to see these opportunities realized.
Collapse
Affiliation(s)
- Yang Xu
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jason C Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia; .,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
5
|
Chen Z, Wu S, Zeng Y, Li X, Wang M, Chen Z, Chen M. The antifibrotic and anti-inflammatory effects of FZHY prescription on the kidney in rats after unilateral ureteral obstruction. Acta Cir Bras 2023; 37:e371003. [PMID: 36629622 PMCID: PMC9829242 DOI: 10.1590/acb371003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 09/13/2022] [Indexed: 01/11/2023] Open
Abstract
PURPOSE To explore the potential impact of traditional Chinese herb FuZhengHuaYuJiangZhuTongLuo recipe (FZHY) on renal interstitial fibrosis (RIF) in chronic kidney disease (CKD) at cellular and molecular levels. METHODS Unilateral ureteral obstruction (UUO) rats were established as the RIF model in vivo. The rats were given intragastric administration with FZHY once a day for consecutive 7, 14 and 21 days, respectively. The renal function parameters and inflammation indicators in kidney tissues were measured using enzyme-linked immunosorbent assay, the CD4+/CD8+ T cells in peripheral blood was detected using flow cytometry, the renal fibrosis degree was estimated using Masson's staining, and the fibrosis-related genes' expression was detected using quantitative polymerase chain reaction, western blotting, and immunohistochemistry analyses. RESULTS FZHY prescription reduced the serum creatinine and blood urea nitrogen, decreased the levels of c-reactive protein, interleukin-1, interleukin-6 and tumor necrosis factor-α in kidney tissues, and increased the ratio of CD4+/CD8+ T cells in peripheral blood. FZHY prescription suppressed the renal tissue fibrosis and reduced the levels of laminin, fibronectin, collagen I and collagen III. CONCLUSIONS FZHY prescription suppressed the renal fibrosis and improved the condition of "Healthy Qi Deficiency and Evil Qi Excess" in rats with UUO, which may provide an effective method for CKD treatment.
Collapse
Affiliation(s)
- Ziwei Chen
- M.M. Chengdu University of Traditional Chinese Medicine – Department of Nephrology – Affiliated Integrated TCM and Western Medicine Hospital of Chengdu – Chengdu Integrated TCM and Western Medicine Hospital – Chengdu First People’s Hospital – Chengdu, China.,Corresponding author:
- (86) 18980880236
| | - Shaobo Wu
- M.M. Chengdu University of Traditional Chinese Medicine – Department of Nephrology – Hospital of Chengdu – Chengdu, China
| | - Yu Zeng
- B.S. Chengdu University of Traditional Chinese Medicine – Department of Clinical Laboratory – Hospital of Chengdu – Chengdu, China
| | - Xueying Li
- M.M. Chengdu University of Traditional Chinese Medicine – Department of Nephrology – Hospital of Chengdu – Chengdu, China
| | - Mengping Wang
- M.M. Chengdu University of Traditional Chinese Medicine – Department of Nephrology – Hospital of Chengdu – Chengdu, China
| | - Zejun Chen
- M.D. Chengdu University of Traditional Chinese Medicine – Department of Nephrology – Affiliated Integrated TCM and Western Medicine Hospital of Chengdu – Chengdu Integrated TCM and Western Medicine Hospital – Chengdu First People’s Hospital – Chengdu, China
| | - Ming Chen
- M.M. Chengdu University of Traditional Chinese Medicine – Department of Nephrology – Hospital of Chengdu – Chengdu, China.,Corresponding author:
- (86) 18980880236
| |
Collapse
|
6
|
Association of Netrin 1 with hsCRP in Subjects with Obesity and Recent Diagnosis of Type 2 Diabetes. Curr Issues Mol Biol 2022; 45:134-140. [PMID: 36661496 PMCID: PMC9857863 DOI: 10.3390/cimb45010010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/07/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Netrin 1 (Ntn1) is a cell migration protein with an anti-inflammatory effect, which may play a key role in the pathological development of type 2 diabetes (T2D). In this study, we evaluate the relationships between the serum concentrations of Ntn1, glucose, and high-sensitivity C-reactive Protein (hsCRP). We carried out a cross-sectional study including 90 individuals divided into three groups (n = 30): healthy subjects, individuals with obesity without glucose alterations, and individuals with newly diagnosed T2D. Serum concentrations of Ntn1 and hs-CRP were determined by enzyme-linked immunosorbent assay (ELISA). The serum concentration of Ntn1 was higher in individuals with newly diagnosed T2D (0.33 ± 0.22 ng/mL), in comparison to healthy subjects and individuals with obesity (0.13 ± 0.06 and 0.15 ± 0.07 ng/mL, respectively). In addition, we observed a positive association between the levels of Ntn1 and hsCRP (rho = 0.443; p < 0.001) as well as with serum glucose (rho = −0.110; p = 0.05). The serum concentration of Ntn1 was higher in individuals with T2D, in comparison with the other groups in this study, and presented a positive correlation with hsCRP. Therefore, Ntn1 can be considered a promising risk biomarker and a potential therapeutic target for T2D.
Collapse
|
7
|
Chen S, Zhou M, Ying X, Zhou C. Ellagic acid protects rats from chronic renal failure via MiR-182/FOXO3a axis. Mol Immunol 2021; 138:150-160. [PMID: 34428620 DOI: 10.1016/j.molimm.2021.08.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 01/20/2023]
Abstract
Studies showed that ellagic acid (EA) can significantly improve kidney function, but the renal-protective effects of EA and the potential mechanism require adequate elucidation. This study investigated the mechanisms of EA in chronic renal failure (CRF) injury. A rat model of CRF was established by 5/6 nephrectomy. The body weight, urine volume and urine protein content of the rat model of CRF with EA treatment (0/20/40 mg/kg/day) were recorded. Hematoxylin&eosin (H&E) staining, Masson staining and TUNEL were used for histopathological observation. Serum levels of creatinine value, blood urea nitrogen, superoxide dismutase, glutathione, malondialdehyde, tumor necrosis factor-α, interleukin-6 and intercellular cell adhesion molecule-1 were determined using enzyme-linked immunosorbent assay (ELISA) kits. The expressions of genes involved in CRF damage were detected by quantitative real-time PCR (qRT-PCR) and western blot. The relationships among EA, miR-182 and FOXO3a were verified by TargetScan 7.2, dual-luciferase assay and rescue experiments. In this study, EA treatment significantly increased the body weight, but reduced urination and urine protein content, renal tissue damage, collagen deposition, inflammation and the contents of serum creatinine (Scr), blood urea nitrogen (BUN), and malondialdehyde (MDA), and improved the antioxidant capacity of CRF rats. Moreover, EA treatment inhibited miR-182, TGF-β1, fibronectin and Bax levels, and promoted those of FOXO3a and Bcl-2 in CRF rats. Additionally, miR-182 specifically targeted FOXO3a, and effectively reduced the renal-protective effect of EA. Further research found that overexpressed FOXO3a partially reversed the inhibitory effect of miR-182 on CRF rats. Our results suggest that EA might reduce CRF injury in rats via miR-182/FOXO3a.
Collapse
Affiliation(s)
- Siqi Chen
- Department of Nephrology, The Affiliated People's Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China
| | - Meiyang Zhou
- Department of Nephrology, The Affiliated People's Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China
| | - Xuxia Ying
- Department of Intensive Care Unit, The Affiliated People's Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China
| | - Canxin Zhou
- Department of Nephrology, The Affiliated People's Hospital, Ningbo University, Ningbo, Zhejiang, 315040, China.
| |
Collapse
|
8
|
Duan L, Woolbright BL, Jaeschke H, Ramachandran A. Late Protective Effect of Netrin-1 in the Murine Acetaminophen Hepatotoxicity Model. Toxicol Sci 2021; 175:168-181. [PMID: 32207522 DOI: 10.1093/toxsci/kfaa041] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Acetaminophen (APAP) overdose-induced acute liver failure is an important clinical problem in the United States and the current antidote N-acetylcysteine, has a short early therapeutic window. Since most patients present late to the clinic, there is need for novel late-acting therapeutic options. Though the neuronal guidance cue netrin-1, has been shown to promote hepatic repair and regeneration during liver ischemia/reperfusion injury, its effect in APAP-induced hepatotoxicity is unknown. In the quest for a late-acting therapeutic intervention in APAP-induced liver injury, we examined the role of netrin-1 in a mouse model of APAP overdose. Male C57BL/6J mice were cotreated with exogenous netrin-1 or vehicle control, along with 300 mg/kg APAP and euthanized at 6, 12, and 24 h. Significant elevations in alanine aminotransferase indicative of liver injury were seen in control mice at 6 h and this was not affected by netrin-1 administration. Also, netrin-1 treatment did not influence mitochondrial translocation of phospho-JNK, or peroxynitrite formation indicating that there was no interference with APAP-induced injury processes. Interestingly however, netrin-1 administration attenuated liver injury at 24 h, as seen by alanine aminotransferase levels and histology, at which time significant elevations in the netrin-1 receptor, adenosine A2B receptor (A2BAR) as well as macrophage infiltration was evident. Removal of resident macrophages with clodronate liposomes or treatment with the A2BAR antagonist PSB1115 blocked the protective effects of netrin-1. Thus, our data indicate a previously unrecognized role for netrin-1 in attenuation of APAP hepatotoxicity by enhancing recovery and regeneration, which is mediated through the A2BAR and involves resident liver macrophages.
Collapse
Affiliation(s)
- Luqi Duan
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Benjamin L Woolbright
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160
| |
Collapse
|
9
|
Li S, Wang F, Sun D. The renal microcirculation in chronic kidney disease: novel diagnostic methods and therapeutic perspectives. Cell Biosci 2021; 11:90. [PMID: 34001267 PMCID: PMC8130426 DOI: 10.1186/s13578-021-00606-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 05/06/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic kidney disease (CKD) affects 8–16% of the population worldwide and is characterized by fibrotic processes. Understanding the cellular and molecular mechanisms underpinning renal fibrosis is critical to the development of new therapeutics. Microvascular injury is considered an important contributor to renal progressive diseases. Vascular endothelium plays a significant role in responding to physical and chemical signals by generating factors that help maintain normal vascular tone, inhibit leukocyte adhesion and platelet aggregation, and suppress smooth muscle cell proliferation. Loss of the rich capillary network results in endothelial dysfunction, hypoxia, and inflammatory and oxidative effects and further leads to the imbalance of pro- and antiangiogenic factors, endothelial cell apoptosis and endothelial-mesenchymal transition. New techniques, including both invasive and noninvasive techniques, offer multiple methods to observe and monitor renal microcirculation and guide targeted therapeutic strategies. A better understanding of the role of endothelium in CKD will help in the development of effective interventions for renal microcirculation improvement. This review focuses on the role of microvascular injury in CKD, the methods to detect microvessels and the novel treatments to ameliorate renal fibrosis.
Collapse
Affiliation(s)
- Shulin Li
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China
| | - Fei Wang
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China
| | - Dong Sun
- Department of Nephrology, Affiliated Hospital of Xuzhou Medical University, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, China. .,Department of Internal Medicine and Diagnostics, Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
10
|
Abd El-Khalik SR, Sharaby RM, Nasif E, Hamza MB, Ibrahim RR. Netrin-1 and clusterin: Innovative potential diagnostic biomarkers for early renal damage in β-thalassemia major children. IUBMB Life 2021; 73:800-810. [PMID: 33715293 DOI: 10.1002/iub.2464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 02/17/2021] [Accepted: 02/22/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Children with β-thalassemia major (β-TM) suffer from tubular dysfunction even before the onset of any renal impairment symptoms and/or clinical signs. Therefore, identifying innovative biomarkers allowing early renal damage detection has focused attention. AIM This study aims to preliminary assess Netrin-1(NTN-1) and clusterin (CLU) in β-TM children and explore their possible roles as surrogate noninvasive biomarkers of renal tubular dysfunction. SUBJECTS AND METHODS In this study, 40 β-TM children and 30 healthy children were enrolled. Routine serum and urinary biochemical variables were determined. Urinary NTN-1 and CLU levels were measured using ELISA and their mRNA expression in PBMCs were assayed using real-time PCR. Serum TNF-α, MDA levels and GST activity were measured. RESULTS Urinary NTN-1 and CLU concentrations and mRNA relative expression levels in PBMCs were significantly increased in β-TM children relative to controls. Oxidative stress and inflammatory markers revealed significant elevation in β-TM children compared to controls. The change in these parameters correlated significantly with other renal parameters. ROC curves analysis showed that urinary NTN-1 and CLU levels are of promising diagnostic performance. CONCLUSION Our results suggest that NTN-1 and CLU are qualified as new noninvasive biomarker panels for early detection of renal injury in β-TM children. Moreover, urinary NTN-1 is recommended as a precise one during the clinical practices.
Collapse
Affiliation(s)
- Sarah Ragab Abd El-Khalik
- Medical Biochemistry & Molecular Biology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | - Elham Nasif
- Physiology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | | | - Rowida Raafat Ibrahim
- Medical Biochemistry & Molecular Biology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
11
|
Mehta N, Li R, Zhang D, Soomro A, He J, Zhang I, MacDonald M, Gao B, Krepinsky JC. miR299a-5p promotes renal fibrosis by suppressing the antifibrotic actions of follistatin. Sci Rep 2021; 11:88. [PMID: 33420269 PMCID: PMC7794215 DOI: 10.1038/s41598-020-80199-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 12/15/2020] [Indexed: 01/29/2023] Open
Abstract
Caveolin-1 (cav-1), an integral protein of the membrane microdomains caveolae, is required for synthesis of matrix proteins by glomerular mesangial cells (MC). Previously, we demonstrated that the antifibrotic protein follistatin (FST) is transcriptionally upregulated in cav-1 knockout MC and that its administration is protective against renal fibrosis. Here, we screened cav-1 wild-type and knockout MC for FST-targeting microRNAs in order to identity novel antifibrotic therapeutic targets. We identified that miR299a-5p was significantly suppressed in cav-1 knockout MC, and this was associated with stabilization of the FST 3'UTR. Overexpression and inhibition studies confirmed the role of miR299a-5p in regulating FST expression. Furthermore, the profibrotic cytokine TGFβ1 was found to stimulate the expression of miR299a-5p and, in turn, downregulate FST. Through inhibition of FST, miR299a-5p overexpression augmented, while miR299a-5p inhibition diminished TGFβ1 profibrotic responses, whereas miR299a-5p overexpression re-enabled cav-1 knockout MC to respond to TGFβ1. In vivo, miR299a-5p was upregulated in the kidneys of mice with chronic kidney disease (CKD). miR299a-5p inhibition protected these mice against renal fibrosis and CKD severity. Our data demonstrate that miR299a-5p is an important post-transcriptional regulator of FST, with its upregulation an important pathogenic contributor to renal fibrosis. Thus, miR299a-5p inhibition offers a potential novel therapeutic approach for CKD.
Collapse
Affiliation(s)
- Neel Mehta
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Renzhong Li
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Dan Zhang
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Asfia Soomro
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Juehua He
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Ivan Zhang
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Melissa MacDonald
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Bo Gao
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Joan C Krepinsky
- Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada.
- St. Joseph's Hospital, 50 Charlton Ave East, Rm T3311, Hamilton, ON, L8N 4A6, Canada.
| |
Collapse
|
12
|
Wang Y, Wang Y, Li Y, Lu L, Peng Y, Zhang S, Xia A. Metformin attenuates renal interstitial fibrosis through upregulation of Deptor in unilateral ureteral obstruction in rats. Exp Ther Med 2020; 20:17. [PMID: 32934682 PMCID: PMC7471900 DOI: 10.3892/etm.2020.9144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 01/14/2020] [Indexed: 12/27/2022] Open
Abstract
Renal interstitial fibrosis (RIF) is a common pathological process that accompanies chronic kidney disease (CKD) and that progresses to end-stage renal failure (ESRD). Accumulating evidence has revealed that persistent mammalian target of rapamycin (mTOR) activation in kidneys is closely associated with the occurrence and progression of CKD. The DEP domain-containing mTOR interacting protein (Deptor) is an endogenous negative regulator of mTOR. Metformin can attenuate renal fibrosis in an animal model of diabetic nephropathy. Previous studies demonstrated that metformin can attenuate renal fibrosis in several models of CKD. However, the precise mechanisms of this effect are not well understood. The present study aimed to examine the mechanism of action of metformin on unilateral ureteral obstruction (UUO)-induced RIF in rats in vivo. Sprague-Dawley rats were randomly divided into a sham-operated group, three UUO groups examined at different time points (3, 7 and 14 days after UUO surgery), and three metformin-treated groups, treated with three different concentrations of metformin. The metformin-treated groups were administered metformin orally every day for 14 consecutive days following surgery. The protein expression levels of Deptor, α-smooth muscle actin (α-SMA), phosphorylated (p-)mTOR, p-ribosomal protein S6 kinase (p-p70S6K) and CD68 were assessed. The present results suggested that, following UUO, there was a significant reduction of Deptor expression, and an increase in collagen deposition in the extracellular matrix over time, accompanied by an increased expression of several proteins including CD68, α-SMA, p-mTOR and p-p70S6K. Notably, metformin treatment reversed these effects. In conclusion, the present results suggested that metformin attenuated RIF of UUO rats, and the mechanism of action was found to be associated with the increase in Deptor expression and inhibition of the mTOR/p70S6K pathway in the kidneys of UUO rats.
Collapse
Affiliation(s)
- Yanxia Wang
- Department of Pharmacology, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Yan Wang
- Department of Pharmacy, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Yong Li
- Department of Gastrointestinal Surgery, Xuzhou Central Hospital, Xuzhou, Jiangsu 221006, P.R. China
| | - Linghong Lu
- Department of Pharmacology, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Yingxian Peng
- Department of Pharmacology, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Shu Zhang
- Department of Pharmacology, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| | - Anzhou Xia
- Department of Pharmacology, Xuzhou Medical University, Xuzhou, Jiangsu 221000, P.R. China
| |
Collapse
|
13
|
Abstract
Endothelial cells and mesenchymal cells are two different cell types with distinct morphologies, phenotypes, functions, and gene profiles. Accumulating evidence, notably from lineage-tracing studies, indicates that the two cell types convert into each other during cardiovascular development and pathogenesis. During heart development, endothelial cells transdifferentiate into mesenchymal cells in the endocardial cushion through endothelial-to-mesenchymal transition (EndoMT), a process that is critical for the formation of cardiac valves. Studies have also reported that EndoMT contributes to the development of various cardiovascular diseases, including myocardial infarction, cardiac fibrosis, valve calcification, endocardial elastofibrosis, atherosclerosis, and pulmonary arterial hypertension. Conversely, cardiac fibroblasts can transdifferentiate into endothelial cells and contribute to neovascularization after cardiac injury. However, progress in genetic lineage tracing has challenged the role of EndoMT, or its reversed programme, in the development of cardiovascular diseases. In this Review, we discuss the caveats of using genetic lineage-tracing technology to investigate cell-lineage conversion; we also reassess the role of EndoMT in cardiovascular development and diseases and elaborate on the molecular signals that orchestrate EndoMT in pathophysiological processes. Understanding the role and mechanisms of EndoMT in diseases will unravel the therapeutic potential of targeting this process and will provide a new paradigm for the development of regenerative medicine to treat cardiovascular diseases.
Collapse
|
14
|
Bian X, Bai Y, Su X, Zhao G, Sun G, Li D. Knockdown of periostin attenuates 5/6 nephrectomy‐induced intrarenal renin–angiotensin system activation, fibrosis, and inflammation in rats. J Cell Physiol 2019; 234:22857-22873. [PMID: 31127625 DOI: 10.1002/jcp.28849] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 04/30/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022]
Affiliation(s)
- Xiaohui Bian
- Department of Nephrology Shengjing Hospital of China Medical University Shenyang P.R. China
| | - Yu Bai
- Department of Nephrology Shengjing Hospital of China Medical University Shenyang P.R. China
| | - Xiaoxiao Su
- Department of Nephrology Shengjing Hospital of China Medical University Shenyang P.R. China
| | - Guifeng Zhao
- Research Center Shengjing Hospital of China Medical University Shenyang P.R. China
| | - Guangping Sun
- Department of Nephrology Shengjing Hospital of China Medical University Shenyang P.R. China
| | - Detian Li
- Department of Nephrology Shengjing Hospital of China Medical University Shenyang P.R. China
| |
Collapse
|
15
|
Novel role of Snail 1 in promoting tumor neoangiogenesis. Biosci Rep 2019; 39:BSR20182161. [PMID: 30975732 PMCID: PMC6509058 DOI: 10.1042/bsr20182161] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 03/14/2019] [Accepted: 03/24/2019] [Indexed: 12/13/2022] Open
Abstract
Snail1 plays an important role in epithelial to mesenchymal transition (EMT) during tumor metastasis; however, whether Snai1 potentiates the process of neoangiogenesis is completely unknown. In the present study, tube formation assay was used to evaluate neoangiogenesis in vitro The expression of Snai1 and other pro-neoangiogenic factors was measured by quantitative real time PCR. Tumor derived endothelial cells (TDECs) were stimulated with fibroblast growth factor 1 (FGF1) or VEGF and formed more tubes compared with untreated, whereas cells treated with Sulforaphane had less tube formation. Silencing SNAI1 significantly attenuated tube formation accompanied by decreased CD31, CD34, and VWF expression in TDECs compared with control. In contrast, overexpression of Snai1 led to more CD31, CD34, and VWF expression and tube formation. To determine if the observed effects of SNAI1 on tube formation was a global phenomenon, the same assay was conducted in normal mesenchymal stem cells (MSCs). SNAI1 silencing did not have any effect on tube formation in MSCs. The expression of TIMP2, ENG, and HIF1A was up-regulated 3-fold or higher after silencing SNAI1, and ID1, VEGFA, PLG, LECT1, HPSE were shown down-regulated. Taken together, our study elucidates an important role of EMT inducer Snai1 in regulating tumor neoangiogenesis, suggesting a potential therapeutic target for overcoming tumor EMT.
Collapse
|
16
|
Bezhaeva T, Geelhoed WJ, Wang D, Yuan H, van der Veer EP, Alem CMAV, Damanik FFR, Qiu X, Zonneveld AJV, Moroni L, Li S, Rotmans JI. Contribution of bone marrow-derived cells to in situ engineered tissue capsules in a rat model of chronic kidney disease. Biomaterials 2018; 194:47-56. [PMID: 30580195 DOI: 10.1016/j.biomaterials.2018.12.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/12/2022]
Abstract
Tissue engineered blood vessels (TEBVs) hold great promise for clinical use in patients with end stage renal disease (ESRD) requiring vascular access for hemodialysis. A promising way to make TEBVs is to exploit foreign body response (FBR) of polymeric rods used as templates. However, since the FBR predominantly involves bone-marrow (BM) derived cells and ESRD coincides with impaired function of BM, it is important to assess the generation of TEBVs in conditions of renal failure. To this end, we implanted polymer rods in the subcutis of rats after BM-transplantation with GFP-labeled BM cells in a model of chronic kidney disease (CKD). At 3 weeks after implantation, rods were encapsulated by tissue capsule (TC) composed of collagen, myofibroblasts and macrophages. On average, 13% of CD68+ macrophages were GFP+, indicating BM origin. Macrophage-to-myofibroblasts differentiation appeared to play an important role in TC formation as 26% of SMA+/GFP+ myofibroblasts co-expressed the macrophage marker CD68. Three weeks after rod implantation, the cellular response changed towards tissue repair, characterized by 40% increase in CD68+/CD163+ repair associated macrophages and 95% increase in TGFβ and IL10 gene expression as compared to TCs harvested at 1 week. These results show that both BM derived and tissue resident cells, contribute to TC formation, whereas macrophages serve as precursors of myofibroblasts in mature TCs. Finally, the presence of CKD did not significantly alter the process of TC formation, which holds the potential to support our approach for future clinical use in ESRD patients.
Collapse
Affiliation(s)
- Taisiya Bezhaeva
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
| | - Wouter J Geelhoed
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
| | - Dong Wang
- Department of Bioengineering and Department of Medicine, University of California, Los Angeles, USA; Department of Bioengineering, University of California, Berkeley, USA
| | - Haoyong Yuan
- Department of Bioengineering and Department of Medicine, University of California, Los Angeles, USA
| | - Eric P van der Veer
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
| | - Carla M A van Alem
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
| | - Febriyani F R Damanik
- MERLN Institute for Technology Inspired Regenerative Medicine, Complex Tissue Regeneration, Maastricht University, the Netherlands
| | - Xuefeng Qiu
- Department of Bioengineering and Department of Medicine, University of California, Los Angeles, USA; Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Anton-Jan van Zonneveld
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands
| | - Lorenzo Moroni
- MERLN Institute for Technology Inspired Regenerative Medicine, Complex Tissue Regeneration, Maastricht University, the Netherlands
| | - Song Li
- Department of Bioengineering and Department of Medicine, University of California, Los Angeles, USA; Department of Bioengineering, University of California, Berkeley, USA; Department of Medicine, University of California, Los Angeles, USA
| | - Joris I Rotmans
- Department of Internal Medicine, Leiden University Medical Center, the Netherlands; Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, the Netherlands.
| |
Collapse
|
17
|
Man S, Sanchez Duffhues G, Ten Dijke P, Baker D. The therapeutic potential of targeting the endothelial-to-mesenchymal transition. Angiogenesis 2018; 22:3-13. [PMID: 30076548 PMCID: PMC6510911 DOI: 10.1007/s10456-018-9639-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 07/27/2018] [Indexed: 12/20/2022]
Abstract
Endothelial cells (ECs) have been found to be capable of acquiring a mesenchymal phenotype through a process known as endothelial-to-mesenchymal transition (EndMT). First seen in the developing embryo, EndMT can be triggered postnatally under certain pathological conditions. During this process, ECs dedifferentiate into mesenchymal stem-like cells (MSCs) and subsequently give rise to cell types belonging to the mesoderm lineage. As EndMT contributes to a multitude of diseases, pharmacological modulation of the signaling pathways underlying EndMT may prove to be effective as a therapeutic treatment. Additionally, EndMT in ECs could also be exploited to acquire multipotent MSCs, which can be readily re-differentiated into various distinct cell types. In this review, we will consider current models of EndMT, how manipulation of this process might improve treatment of clinically important pathologies and how it could be harnessed to advance regenerative medicine and tissue engineering.
Collapse
Affiliation(s)
- Shirley Man
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| | - Gonzalo Sanchez Duffhues
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands.
| | - David Baker
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Einthovenweg 20, 2300 RC, Leiden, The Netherlands
| |
Collapse
|
18
|
Yimer EM, Zewdie KA, Hishe HZ. Netrin as a Novel Biomarker and Its Therapeutic Implications in Diabetes Mellitus and Diabetes-Associated Complications. J Diabetes Res 2018; 2018:8250521. [PMID: 30320139 PMCID: PMC6167572 DOI: 10.1155/2018/8250521] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/14/2018] [Accepted: 09/04/2018] [Indexed: 12/16/2022] Open
Abstract
Diabetes is a multifactorial metabolic syndrome and is one of the shared long-lasting illnesses globally. It is linked to long-term microvascular and macrovascular complications that contribute to disability, compromised quality of life, and reduction in lifespan, which eventually leads to death. This disease is not only incurring significant economic burden but also adversely affects the patients, caregivers, communities, and the society at large. The interruption of diabetes progress and its complications is a primary focus of scientific communities. In spite of various diagnostic modalities for diabetes, there is a limited marker to investigate the risk and progress of its complications. Netrin has recently received more attention as a biomarker of diabetes and a broader range of long-term complication. Therefore, the impetus of this review is to exhaustively discuss the role of Netrin as a potential biomarker and its therapeutic implication in diabetes and diverse sets of microvascular and macrovascular complications of diabetes. It also discourses the possible mechanisms of Netrin for the said pharmacological effect for a better understanding of the development and progression of diabetes and its complications in relation to this protein. It enables protective measures to be applied at the subclinical stage and the responses to preventive or therapeutic measures to be scrutinized. Besides, it might also facilitate the appraisal of novel therapeutic options for diabetes and various complications through modifying the endogenous Netrin and provide surrogate endpoints for intervention.
Collapse
Affiliation(s)
- Ebrahim M. Yimer
- Department of Pharmacology and Toxicology, College of Health Sciences, Mekelle University, Ethiopia
| | - Kaleab Alemayehu Zewdie
- Department of Pharmacology and Toxicology, College of Health Sciences, Mekelle University, Ethiopia
| | - Hailemichael Zeru Hishe
- Department of Pharmacology and Toxicology, College of Health Sciences, Mekelle University, Ethiopia
| |
Collapse
|