1
|
Phongpao K, Kheansaard W, Pholngam N, Sriwantana T, Paiboonsukwong K, Fucharoen S, Pattanapanyasat K, Sibmooh N, Chaichompoo P, Svasti S. Extracellular vesicles modulate endothelial nitric oxide production in patients with β‑thalassaemia/HbE. Biomed Rep 2025; 22:79. [PMID: 40093508 PMCID: PMC11904759 DOI: 10.3892/br.2025.1957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Thrombosis is a significant complication in patients with β-thalassaemia/haemoglobin E (HbE), particularly in splenectomised patients. The endothelium is a key regulator of vascular haemostasis and homeostasis, through the secretion of various regulatory molecules. Nitric oxide (NO), produced by endothelial cells (ECs), regulates vascular functions by acting as a potent vasodilator and an inhibitor of platelet activation. Decreased NO bioavailability, a marker of vascular dysfunction, could be a contributing factor leading to thrombosis. Microparticles or medium extracellular vesicles (mEVs) are associated with thrombosis and vasculopathy in various diseases. Furthermore, elevated levels of mEVs have been observed in splenectomised patients with β-thalassaemia/HbE and could induce the expression of coagulation proteins, inflammatory cytokines and adhesion molecules in ECs. However, the effects of mEVs on NO regulation by ECs is currently unclear. In the present study, mEVs obtained from splenectomised patients with β-thalassaemia/HbE had significantly decreased NO production in human pulmonary artery ECs without affecting endothelial nitric oxide synthase expression or phosphorylation. Decreased NO production was attributed to increased haemoglobin levels in mEVs from splenectomised patients, leading to enhanced NO scavenging. These findings highlight a mechanism whereby haemoglobin-carrying mEVs directly scavenge NO, contributing to vascular dysfunction in β-thalassaemia/HbE disease.
Collapse
Affiliation(s)
- Kunwadee Phongpao
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Wasinee Kheansaard
- Department of Clinical Microscopy, Faculty of Medical Technology, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Nuttanan Pholngam
- Graduate Program in Molecular Medicine, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Thanaporn Sriwantana
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| | - Kittiphong Paiboonsukwong
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Kovit Pattanapanyasat
- Center of Excellence for Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Nuthawut Sibmooh
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan 10540, Thailand
| | - Pornthip Chaichompoo
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Saovaros Svasti
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom 73170, Thailand
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
2
|
Sandrine MNY, Michel BG, Bibi-Farouck AO, Ngo Bahebeck P, Emmanuel OP, Claude BD, Bidingha A Goufani R, Carlos TY, Dupon AAB, Fils EA, Cynthia BYP, Ulrich NTJ, Désiré DDP. In silico molecular docking and predictive ADME properties, in vitro antioxidant scavenging capacities, and in vivo pharmacological activities to study the potential of Pterocarpus mildbraedii's Harms (Fabaceae) in preventing vaginal dysbiosis and risk factors for cardiovascular disease in an estropause rat model. Fitoterapia 2025; 183:106496. [PMID: 40147737 DOI: 10.1016/j.fitote.2025.106496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 02/03/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Vaginal dysbiosis (VaD) is a common issue among menopausal women, who are particularly susceptible to cardiovascular disease (CVD). Pterocarpus sp. are known to induce estrogen-like activities, which are the key pathways for menopause-related disorders. This study aims to evaluate the potential of Pterocarpus mildbraedii water extract on VaD and CVD risk factors using an estropause (EP) rat model. Furthermore, predictive ADME properties and molecular docking with target proteins were assessed to develop alternative medicinal treatments for menopause. The secondary metabolites in P. mildbraedii water extract (Pm) were analyzed using UHPLC-MS and quantitative phytochemistry methods. The extract's ability to scavenge free radicals was evaluated using DPPH, ABTS, and FRAP tests. Molecular docking assessed the extract's binding ability to various receptors. SwissADME and Molinspiration were utilized to predict its pharmacokinetic and bioactivity properties. Subsequently, the therapeutic potential of Pm was assessed in rats, focusing on its estrogen-like, eubiotic, and cardioprotective activities. UHPLC-MS enables us to identify several compounds. Predictive ADME analyses have indicated that most compounds comply with Lipinski's Rule of Five for oral drugs. Additionally, they inhibit CYP1A2 and bind to several receptors and enzymes through conventional hydrogen bonding. In rats, ovariectomy-induced EP reduced glycogen levels and vaginal lactic acid and decreased in the population of Lactobacillus spp., which is characteristic of VaD. This condition also increases CVD risk factors. Overall, this study underscores the potential of Pterocarpus mildbraedii in preventing VaD and CVD risk factors related to hypoestrogenism. This extract positions itself as a promising alternative treatment for menopause-related disorders.
Collapse
Affiliation(s)
- Mengue Ngadena Yolande Sandrine
- Department of Animal Biology and Physiology, Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon; Neurosciences Axis, Laboratory of Development and Maldevelopment, Department of Psychology, Faculty of Arts, Letters, and Social Science, University of Yaoundé 1, P.O. 755, Yaoundé, Cameroon.
| | - Bindzi Georges Michel
- Department of Animal Biology and Physiology, Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Aboubakar Oumarou Bibi-Farouck
- Department of Physiological Sciences and Biochemistry, Faculty of Medicine and Biomedical Sciences, University of Garoua, P.O. Box: 317, Garoua, Cameroon
| | - Pierrette Ngo Bahebeck
- Institute of Agricultural Research for Development (IRAD), P.O. BOX 2067, Yaoundé, Cameroon
| | - Owona Pascal Emmanuel
- Department of Animal Biology and Physiology, Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Bilanda Danielle Claude
- Department of Animal Biology and Physiology, Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Ronald Bidingha A Goufani
- Department of Animal Biology and Physiology, Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Tcheutchoua Yannick Carlos
- Department of Animal Biology and Physiology, Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Ambamba Akamba Bruno Dupon
- Department of Biochemistry, Faculty of Science, University of Yaoundé 1, P. O. Box 812, Yaoundé, Cameroon; Center of Nutrition and Functional Foods, P.O. Box 8024, Yaoundé, Cameroon
| | - Ella Armand Fils
- Department of Biochemistry, Faculty of Science, University of Yaoundé 1, P. O. Box 812, Yaoundé, Cameroon
| | - Bouguem Yandja Paule Cynthia
- Department of Animal Biology and Physiology, Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Ngueuko Talla Jude Ulrich
- Department of Animal Biology and Physiology, Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| | - Dzeufiet Djomeni Paul Désiré
- Department of Animal Biology and Physiology, Laboratory of Animal Physiology, Faculty of Science, University of Yaoundé 1, P.O. Box 812, Yaoundé, Cameroon
| |
Collapse
|
3
|
Zheng X, Huang R, Yin L, Yao M, Chu J, Yang F, Dong Y, Zhao M, Ma T. Injectable antioxidant hyaluronan/chitosan hydrogel as a platelet-rich plasma and stem cell carrier to promote endometrial regeneration and fertility restoration. Acta Biomater 2025; 195:201-215. [PMID: 39894327 DOI: 10.1016/j.actbio.2025.01.062] [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: 10/24/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Severe damage to the uterine endometrium can lead to thin endometrium and intrauterine adhesions (IUAs), resulting in infertility or complications during pregnancy. Therapies utilizing mesenchymal stem cells (MSCs) and platelet-rich plasma (PRP) represent promising strategies for restoring thin endometrium. However, the low homing rate and functionality of transplanted cells, along with the rapid release of PRP growth factors, limit their therapeutic efficacy. In this study, we developed an in-situ formable and redox-responsive hydrogel composed of thiolated hyaluronan (tHA) and thiolated chitosan (tChi) (tHA-tChi) for encapsulating PRP and mouse adipose-derived stem cells (ADSCs). Our results demonstrate that the tHA-tChi hydrogel exhibits appropriate swelling, injectability, self-healing, and antioxidant properties, alongside a sustained release of PRP growth factors. In vitro experiments indicated that the PRP and ADSCs encapsulated within the hydrogel (ADSCs/tHA-tChi/PRP) stimulated angiogenesis in endothelial cells. In a mouse model of thin endometrium, the ADSCs/tHA-tChi/PRP significantly enhanced endometrial regeneration, as evidenced by increased endometrial thickness and reduced fibrosis. This improvement markedly enhanced endometrial receptivity and pregnancy rates in damaged endometria, correlating with increased angiogenesis and endometrial cell proliferation via activation of the VEGF/AKT/BAD pathway, as shown by Western blotting assays. Overall, the combination of antioxidant hydrogel, PRP, and ADSCs demonstrates promising potential for promoting endometrial regeneration and restoring fertility, offering new minimally invasive therapeutic options for endometrial diseases. STATEMENT OF SIGNIFICANCE: This research presents a potent approach to the treatment of thin endometrium, employing an injectable, biodegradable and antioxidant hydrogel comprising thiolated hyaluronic acid (tHA) and thiolated chitosan (tChi). The antioxidant capacity of the hydrogel improves the oxidative microenvironment of the injured uterus, while the hydrogel is designed to release adipose-derived stem cells (ADSCs) and growth factors from platelet-rich plasma (PRP) sustainably, promoting tissue regeneration by enhancing angiogenesis and endometrium cell proliferation. Demonstrated efficacy in a mouse model of thin endometrium indicates its great potential to significantly improve fertility restoration treatments. The administration of antioxidant hydrogel containing ADSCs and PRP represents a promising therapeutic strategy for patients with endometrial disease.
Collapse
Affiliation(s)
- Xiudan Zheng
- Department of Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, PR China; Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, PR China
| | - Rui Huang
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, PR China
| | - Lanlan Yin
- Department of Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, PR China
| | - Meihua Yao
- Department of Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, PR China
| | - Jiaqi Chu
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, PR China
| | - Fengkai Yang
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, PR China; Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, PR China
| | - Yeying Dong
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, PR China; Spinal Surgery, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, PR China
| | - Mingyan Zhao
- Stem Cell Research and Cellular Therapy Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, PR China.
| | - Tianzhong Ma
- Department of Reproductive Medicine Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, 524001, PR China.
| |
Collapse
|
4
|
Li K, Guo B, Gu J, Ta N, Gu J, Yu H, Sun M, Han T. Emerging advances in drug delivery systems (DDSs) for optimizing cancer complications. Mater Today Bio 2025; 30:101375. [PMID: 39759851 PMCID: PMC11699619 DOI: 10.1016/j.mtbio.2024.101375] [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: 08/02/2024] [Revised: 11/13/2024] [Accepted: 11/29/2024] [Indexed: 01/07/2025] Open
Abstract
The management and treatment of tumor complications pose continuous challenges due to the inherent complexity. However, the advent of drug delivery systems (DDSs) brings promising opportunities to address the tumor complications using innovative technological approaches. This review focuses on common oncological complications, including cancer thrombosis, malignant serous effusion, tumor-associated infections, cancer pain, and treatment-related complications. Emphasis was placed on the application and potential of DDSs in mitigating and treating these tumor complications, and we delved into the underlying mechanisms of common cancer-associated complications, discussed the limitations of conventional treatments, and outlined the current status and potential development of DDSs for various complications in this review. Moreover, we have discussed the existing challenges in DDSs research, underscoring the need for addressing issues related to biocompatibility and targeting of DDSs, optimizing drug delivery routes, and enhancing delivery efficiency and precision. In conclusion, DDSs offer promising avenues for treating cancer complications, offering the potential for the development of more effective and safer drug delivery strategies, thereby improving the quality of life and survival rates of cancer patients.
Collapse
Affiliation(s)
- Kerui Li
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Bei Guo
- Department of Endocrinology, General Hospital of Northern Theater Command, Shenyang, 110001, China
| | - Junmou Gu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China
| | - Na Ta
- Department of Neurology, Second Affiliated Hospital of Dalian Medical University, Dalian, 116044, China
| | - Jia Gu
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Hao Yu
- Department of Endocrinology, General Hospital of Northern Theater Command, Shenyang, 110001, China
| | - Mengchi Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Tao Han
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, 110001, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| |
Collapse
|
5
|
Hu Z, Cano I, Lei F, Liu J, Bossardi Ramos R, Gordon H, Paschalis EI, Saint-Geniez M, Ng YSE, D'Amore PA. Loss of the Endothelial Glycocalyx Component EMCN Leads to Glomerular Impairment. Circ Res 2025; 136:59-74. [PMID: 39584795 DOI: 10.1161/circresaha.124.325218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND EMCN (endomucin), an endothelial-specific glycocalyx component, was found to be highly expressed by the endothelium of the renal glomerulus. We reported an anti-inflammatory role of EMCN and its involvement in the regulation of VEGF (vascular endothelial growth factor) activity through modulating VEGFR2 (VEGF receptor 2) endocytosis. The goal of this study is to investigate the phenotypic and functional effects of EMCN deficiency using the first global EMCN knockout mouse model. METHODS Global EMCN knockout mice were generated by crossing EMCN-floxed mice with ROSA26-Cre mice. Flow cytometry was used to analyze infiltrating myeloid cells in the kidneys. The ultrastructure of the glomerular filtration barrier was examined by transmission electron microscopy, whereas urinary albumin, creatinine, and total protein levels were analyzed from freshly collected urine samples. Expression and localization of EMCN, EGFP (enhanced green fluorescent protein), CD45 (cluster of differentiation 45), CD31, CD34, podocin, and albumin were examined by immunohistochemistry. Mice were weighed regularly, and their systemic blood pressure was measured using a noninvasive tail-cuff system. Glomerular endothelial cells and podocytes were isolated by fluorescence-activated cell sorting for RNA sequencing. Transcriptional profiles were analyzed to identify differentially expressed genes in both endothelium and podocytes, followed by gene ontology analysis. Protein levels of EMCN, albumin, and podocin were quantified by Western blot. RESULTS The EMCN-/- mice exhibited increased infiltration of CD45+ cells, with an increased proportion of Ly6GhighLy6Chigh myeloid cells and higher VCAM-1 (vascular cell adhesion molecule 1) expression. EMCN-/- mice displayed albuminuria with increased albumin in the Bowman's space compared with the EMCN+/+ littermates. Glomeruli in EMCN-/- mice revealed fused and effaced podocyte foot processes and disorganized endothelial fenestrations. We found no significant difference in blood pressure between EMCN knockout mice and their wild-type littermates. RNA sequencing of glomerular endothelial cells revealed downregulation of cell-cell adhesion and MAPK (mitogen-activated protein kinase)/ERK (extracellular signal-regulated kinase) pathways, along with glycocalyx and extracellular matrix remodeling. In podocytes, we observed reduced VEGF signaling and alterations in cytoskeletal organization. Notably, there was a significant decrease in both mRNA and protein levels of podocin, a key component of the slit diaphragm. CONCLUSION Our study demonstrates a critical role of the endothelial marker EMCN in supporting normal glomerular filtration barrier structure and function by maintaining glomerular endothelial tight junction and homeostasis and podocyte function through endothelial-podocyte crosstalk.
Collapse
Affiliation(s)
- Zhengping Hu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
| | - Issahy Cano
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
| | - Fengyang Lei
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
| | - Jie Liu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
| | - Ramon Bossardi Ramos
- Department of Molecular and Cellular Physiology, Albany Medical Center, NY (R.B.R.)
| | - Harper Gordon
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
| | - Eleftherios I Paschalis
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
| | - Magali Saint-Geniez
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
- Now with Biomedical Research, Novartis, Cambridge, MA (M.S.-G.)
| | - Yin Shan Eric Ng
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
- Now with EyeBiotech Limited, a subsidiary of Merck & Co, Inc, Rahway, NJ (Y.S.E.N.)
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Boston, MA (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.)
- Department of Ophthalmology (Z.H., I.C., F.L., J.L., H.G., E.I.P., M.S.-G., Y.S.E.N., P.A.D.), Harvard Medical School, Boston, MA
- Department of Pathology (P.A.D.), Harvard Medical School, Boston, MA
| |
Collapse
|
6
|
Cronin NM, Dawson LW, DeMali KA. Shear stress-stimulated AMPK couples endothelial cell mechanics, metabolism and vasodilation. J Cell Sci 2024; 137:jcs262232. [PMID: 39513477 PMCID: PMC11795286 DOI: 10.1242/jcs.262232] [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: 04/29/2024] [Accepted: 11/02/2024] [Indexed: 11/15/2024] Open
Abstract
Endothelial cells respond to mechanical force by stimulating cellular signaling, but how these pathways are linked to elevations in cell metabolism and whether metabolism supports the mechanical response remains poorly understood. Here, we show that the application of force to endothelial cells stimulates VE-cadherin to activate liver kinase B1 (LKB1; also known as STK11) and AMP-activated protein kinase (AMPK), a master regulator of energy homeostasis. VE-cadherin-stimulated AMPK increases eNOS (also known as NOS3) activity and localization to the plasma membrane, reinforcement of the actin cytoskeleton and cadherin adhesion complex, and glucose uptake. We present evidence for the increase in metabolism being necessary to fortify the adhesion complex, actin cytoskeleton and cellular alignment. Together, these data extend the paradigm for how mechanotransduction and metabolism are linked to include a connection to vasodilation, thereby providing new insight into how diseases involving contractile, metabolic and vasodilatory disturbances arise.
Collapse
Affiliation(s)
- Nicholas M. Cronin
- Roy J. and Lucille A. Carver College of Medicine at the University of Iowa, Department of Biochemistry and Molecular Biology, 51 Newton Rd, Iowa City, IA 52242, USA
| | - Logan W. Dawson
- Roy J. and Lucille A. Carver College of Medicine at the University of Iowa, Department of Biochemistry and Molecular Biology, 51 Newton Rd, Iowa City, IA 52242, USA
| | - Kris A. DeMali
- Roy J. and Lucille A. Carver College of Medicine at the University of Iowa, Department of Biochemistry and Molecular Biology, 51 Newton Rd, Iowa City, IA 52242, USA
| |
Collapse
|
7
|
Patidar K, Versypt ANF. Logic-Based Modeling of Inflammatory Macrophage Crosstalk with Glomerular Endothelial Cells in Diabetic Kidney Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.04.535594. [PMID: 37066138 PMCID: PMC10104015 DOI: 10.1101/2023.04.04.535594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Diabetic kidney disease is a complication in one out of three patients with diabetes. Aberrant glucose metabolism in diabetes leads to structural and functional damage in glomerular tissue and a systemic inflammatory immune response. Complex cellular signaling is at the core of metabolic and functional derangement. Unfortunately, the mechanism underlying the role of inflammation in glomerular endothelial cell dysfunction during diabetic kidney disease is not fully understood. Mathematical models in systems biology allow the integration of experimental evidence and cellular signaling networks to understand mechanisms involved in disease progression. This study developed a logic-based ordinary differential equations model to study inflammatory crosstalk between macrophages and glomerular endothelial cells during diabetic kidney disease progression using a protein signaling network stimulated with glucose and lipopolysaccharide. This modeling approach reduced the biological parameters needed to study signaling networks. The model was fitted to and validated against available biochemical data from \textit{in vitro} experiments. The model identified mechanisms for dysregulated signaling in macrophages and glomerular endothelial cells during diabetic kidney disease. In addition, the influence of signaling interactions on glomerular endothelial cell morphology through selective knockdown and downregulation was investigated. Simulation results showed that partial knockdown of VEGF receptor 1, PLC-γ, adherens junction proteins, and calcium partially recovered the intercellular gap width between glomerular endothelial cells. These findings contribute to understanding signaling and molecular perturbations that affect the glomerular endothelial cells in the early stage of diabetic kidney disease.
Collapse
|
8
|
Guarino VA, Wertheim BM, Xiao W, Loscalzo J, Zhang Y. Nanoparticle delivery of VEGF and SDF-1α as an approach for treatment of pulmonary arterial hypertension. Pulm Circ 2024; 14:e12412. [PMID: 39380978 PMCID: PMC11459680 DOI: 10.1002/pul2.12412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 10/10/2024] Open
Abstract
Endothelial dysfunction is an underlying mechanism for the development of pulmonary arterial hypertension (PAH). Vascular endothelial growth factor (VEGF) and stromal cell-derived factor-1α (SDF) may help repair the dysfunctional endothelium and provide treatment for PAH. To examine this possibility, nanoparticles carrying human recombinant VEGF and SDF (VEGFNP and SDFNP) were aerosolized into the lungs of nude rats at Day 14 after monocrotaline (MCT) injection and analyses were performed at Day 28. The data show that the VEGFNP/SDFNP delivery led to a lower pulmonary arterial pressure and prevented right ventricular hypertrophy in the MCT rats: the right ventricular systolic pressure of the control, MCT, and MCT + VEGFNP/SDFNP treatment groups were 29±2, 70±9, and 44±5 (mean±SD) mmHg, respectively; the pulmonary vascular resistance indices of the groups were 0.6±0.3, 3.2±0.7, and 1.7±0.5, respectively; and the Fulton indices [-RV/(LV + Septum)] were 0.22±0.01, 0.44±0.07, and 0.23±0.02, respectively. The VEGFNP/SDFNP delivery delayed the thickening of distal pulmonary vessels: the number of nearly occluded vessels in a whole lung section from the MCT and MCT + VEGFNP/SDFNP groups were 46±12 and 2±3, respectively. Gene expression analysis of the endothelial cell markers, VE-cadherin, KDR, BMPR2, and eNOS, and smooth cell markers, SM-MHC and α-SMA, indicated significant loss of distal pulmonary vessels in the MCT- treated rats. VEGFNP/SDFNP delivery did not recover the loss, but significantly increased eNOS and decreased α-SMA expression in the MCT-treated lungs. Thus, the therapeutic effect of VEGFNP/SDFNP may be mediated by improving/repairing endothelial function in the PAH lungs.
Collapse
Affiliation(s)
| | | | - Wusheng Xiao
- Harvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
| | - Joseph Loscalzo
- Harvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
| | - Ying‐Yi Zhang
- Harvard Medical SchoolBrigham and Women's HospitalBostonMassachusettsUSA
| |
Collapse
|
9
|
Pandian K, Huang L, Junaid A, Harms A, van Zonneveld AJ, Hankemeier T. Tracer-based metabolomics for profiling nitric oxide metabolites in a 3D microvessels-on-chip model. FASEB J 2024; 38:e70005. [PMID: 39171967 DOI: 10.1096/fj.202400553r] [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: 03/12/2024] [Revised: 06/19/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Endothelial dysfunction, prevalent in cardiovascular diseases (CVDs) and linked to conditions like diabetes, hypertension, obesity, renal failure, or hypercholesterolemia, is characterized by diminished nitric oxide (NO) bioavailability-a key signaling molecule for vascular homeostasis. Current two-dimensional (2D) in vitro studies on NO synthesis by endothelial cells (ECs) lack the crucial laminar shear stress, a vital factor in modulating the NO-generating enzyme, endothelial nitric oxide synthase (eNOS), under physiological conditions. Here we developed a tracer-based metabolomics approach to measure NO-specific metabolites with mass spectrometry (MS) and show the impact of fluid flow on metabolic parameters associated with NO synthesis using 2D and 3D platforms. Specifically, we tracked the conversion of stable-isotope labeled NO substrate L-Arginine to L-Citrulline and L-Ornithine to determine eNOS activity. We demonstrated clear responses in human coronary artery endothelial cells (HCAECs) cultured with 13C6, 15N4-L-Arginine, and treated with eNOS stimulator, eNOS inhibitor, and arginase inhibitor. Analysis of downstream metabolites, 13C6, 15N3 L-Citrulline and 13C5, 15N2 L-Ornithine, revealed distinct outcomes. Additionally, we evaluated the NO metabolic status in static 2D culture and 3D microvessel models with bidirectional and unidirectional fluid flow. Our 3D model exhibited significant effects, particularly in microvessels exposed to the eNOS stimulator, as indicated by the 13C6, 15N3 L-Citrulline/13C5, 15N2 L-Ornithine ratio, compared to the 2D culture. The obtained results indicate that the 2D static culture mimics an endothelial dysfunction status, while the 3D model with a unidirectional fluid flow provides a more representative physiological environment that provides a better model to study endothelial dysfunction.
Collapse
Affiliation(s)
- Kanchana Pandian
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Luojiao Huang
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Abidemi Junaid
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Amy Harms
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Thomas Hankemeier
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| |
Collapse
|
10
|
Gaydarski L, Dimitrova IN, Stanchev S, Iliev A, Kotov G, Kirkov V, Stamenov N, Dikov T, Georgiev GP, Landzhov B. Unraveling the Complex Molecular Interplay and Vascular Adaptive Changes in Hypertension-Induced Kidney Disease. Biomedicines 2024; 12:1723. [PMID: 39200188 PMCID: PMC11351430 DOI: 10.3390/biomedicines12081723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/25/2024] [Accepted: 07/31/2024] [Indexed: 09/02/2024] Open
Abstract
Angiogenesis, the natural mechanism by which fresh blood vessels develop from preexisting ones, is altered in arterial hypertension (AH), impacting renal function. Studies have shown that hypertension-induced renal damage involves changes in capillary density (CD), indicating alterations in vascularization. We aimed to elucidate the role of the apelin receptor (APLNR), neuronal nitric oxide synthase (nNOS), and vascular endothelial growth factor (VEGF) in hypertension-induced renal damage. We used two groups of spontaneously hypertensive rats aged 6 and 12 months, representing different stages of AH, and compared them to age-matched normotensive controls. The kidney tissue samples were prepared through a well-established protocol. All data analysis was conducted with a dedicated software program. APLNR was localized in tubular epithelial cells and the endothelial cells of the glomeruli, with higher expression in older SHRs. The localization of nNOS and VEGF was similar. The expression of APLNR and nNOS increased with AH progression, while VEGF levels decreased. CD was lower in young SHRs compared to controls and decreased significantly in older SHRs in comparison to age-matched controls. Our statistical analysis revealed significant differences in molecule expression between age groups and varying correlations between the expression of the three molecules and CD.
Collapse
Affiliation(s)
- Lyubomir Gaydarski
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (L.G.); (S.S.); (A.I.); (N.S.); (B.L.)
| | - Iva N. Dimitrova
- Department of Cardiology, University Hospital “St. Ekaterina”, Medical University of Sofia, 1431 Sofia, Bulgaria;
| | - Stancho Stanchev
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (L.G.); (S.S.); (A.I.); (N.S.); (B.L.)
| | - Alexandar Iliev
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (L.G.); (S.S.); (A.I.); (N.S.); (B.L.)
| | - Georgi Kotov
- Department of Rheumatology, Clinic of Rheumatology, University Hospital “St. Ivan Rilski”, Medical Faculty, Medical University of Sofia, 1612 Sofia, Bulgaria;
| | - Vidin Kirkov
- Department of Health Policy and Management, Faculty of Public Health “Prof. Dr. Tzekomir Vodenicharov”, Medical University of Sofia, 1431 Sofia, Bulgaria;
| | - Nikola Stamenov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (L.G.); (S.S.); (A.I.); (N.S.); (B.L.)
| | - Tihomir Dikov
- Department of General and Clinical Pathology, Medical University of Sofia, 1431 Sofia, Bulgaria;
| | - Georgi P. Georgiev
- Department of Orthopedics and Traumatology, University Hospital Queen Giovanna-ISUL, Medical University of Sofia, 1527 Sofia, Bulgaria
| | - Boycho Landzhov
- Department of Anatomy, Histology and Embryology, Medical University of Sofia, 1431 Sofia, Bulgaria; (L.G.); (S.S.); (A.I.); (N.S.); (B.L.)
| |
Collapse
|
11
|
Hu Z, Cano I, Lei F, Liu J, Ramos RB, Gordon H, Paschalis EI, Saint-Geniez M, Ng YSE, D'Amore PA. Deletion of the endothelial glycocalyx component endomucin leads to impaired glomerular structure and function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603749. [PMID: 39071302 PMCID: PMC11275787 DOI: 10.1101/2024.07.16.603749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Background Endomucin (EMCN), an endothelial-specific glycocalyx component, was found to be highly expressed by the endothelium of the renal glomerulus. We reported an anti-inflammatory role of EMCN and its involvement in the regulation of vascular endothelial growth factor (VEGF) activity through modulating VEGF receptor 2 (VEGFR2) endocytosis. The goal of this study is to investigate the phenotypic and functional effects of EMCN deficiency using the first global EMCN knockout mouse model. Methods Global EMCN knockout mice were generated by crossing EMCN-floxed mice with ROSA26-Cre mice. Flow cytometry was employed to analyze infiltrating myeloid cells in the kidneys. The ultrastructure of the glomerular filtration barrier was examined by transmission electron microscopy, while urinary albumin, creatinine, and total protein levels were analyzed from freshly collected urine samples. Expression and localization of EMCN, EGFP, CD45, CD31, CD34, podocin, albumin, and α-smooth muscle actin were examined by immunohistochemistry. Mice were weighed regularly, and their systemic blood pressure was measured using a non-invasive tail-cuff system. Glomerular endothelial cells and podocytes were isolated by fluorescence-activated cell sorting for RNA-seq. Transcriptional profiles were analyzed to identify differentially expressed genes in both endothelium and podocytes, followed by gene ontology analysis of up- and down-regulated genes. Protein levels of EMCN, albumin, and podocin were quantified by Western blot. Results EMCN -/- mice were viable with no gross anatomical defects in kidneys. The EMCN -/- mice exhibited increased infiltration of CD45 + cells, with an increased proportion of Ly6G high Ly6C high myeloid cells and higher VCAM-1 expression. EMCN -/- mice displayed albuminuria with increased albumin in the Bowman's space compared to the EMCN +/+ littermates. Glomeruli in EMCN -/- mice revealed fused and effaced podocyte foot processes and disorganized endothelial fenestrations. We found no significant difference in blood pressure between EMCN knockout mice and their wild-type littermates. RNA-seq of glomerular endothelial cells revealed downregulation of cell-cell adhesion and MAPK/ERK pathways, along with glycocalyx and extracellular matrix remodeling. In podocytes, we observed reduced VEGF signaling and alterations in cytoskeletal organization. Notably, there was a significant decrease in both mRNA and protein levels of podocin, a key component of the slit diaphragm. Conclusion Our study demonstrates a critical role of the endothelial marker EMCN in supporting normal glomerular filtration barrier structure and function by maintaining glomerular endothelial tight junction and homeostasis and podocyte function through endothelial-podocyte crosstalk.
Collapse
|
12
|
Cronin NM, Dawson LW, DeMali KA. Mechanical activation of VE-cadherin stimulates AMPK to increase endothelial cell metabolism and vasodilation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593171. [PMID: 38798670 PMCID: PMC11118335 DOI: 10.1101/2024.05.09.593171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Endothelia cells respond to mechanical force by stimulating cellular signaling, but how these pathways are linked to elevations in cell metabolism and whether metabolism supports the mechanical response remains poorly understood. Here, we show that application of force to VE-cadherin stimulates liver kinase B1 (LKB1) to activate AMP-activated protein kinase (AMPK), a master regulator of energy homeostasis. VE-cadherin stimulated AMPK increases eNOS activity and localization to the plasma membrane as well as reinforcement of the actin cytoskeleton and cadherin adhesion complex, and glucose uptake. We present evidence for the increase in metabolism being necessary to fortify the adhesion complex, actin cytoskeleton, and cellular alignment. Together these data extend the paradigm for how mechanotransduction and metabolism are linked to include a connection to vasodilation, thereby providing new insight into how diseases involving contractile, metabolic, and vasodilatory disturbances arise.
Collapse
Affiliation(s)
- Nicholas M Cronin
- Roy J. and Lucille A. Carver College of Medicine at the University of Iowa, Department of Biochemistry and Molecular Biology, 51 Newton RD, Iowa City, IA 52242
| | - Logan W Dawson
- Roy J. and Lucille A. Carver College of Medicine at the University of Iowa, Department of Biochemistry and Molecular Biology, 51 Newton RD, Iowa City, IA 52242
| | - Kris A DeMali
- Roy J. and Lucille A. Carver College of Medicine at the University of Iowa, Department of Biochemistry and Molecular Biology, 51 Newton RD, Iowa City, IA 52242
| |
Collapse
|
13
|
Blanot M, Casaroli-Marano RP, Mondéjar-Medrano J, Sallén T, Ramírez E, Segú-Vergés C, Artigas L. Aflibercept Off-Target Effects in Diabetic Macular Edema: An In Silico Modeling Approach. Int J Mol Sci 2024; 25:3621. [PMID: 38612432 PMCID: PMC11011561 DOI: 10.3390/ijms25073621] [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: 01/03/2024] [Revised: 03/08/2024] [Accepted: 03/13/2024] [Indexed: 04/14/2024] Open
Abstract
Intravitreal aflibercept injection (IAI) is a treatment for diabetic macular edema (DME), but its mechanism of action (MoA) has not been completely elucidated. Here, we aimed to explore IAI's MoA and its multi-target nature in DME pathophysiology with an in silico (computer simulation) disease model. We used the Therapeutic Performance Mapping System (Anaxomics Biotech property) to generate mathematical models based on the available scientific knowledge at the time of the study, describing the relationship between the modulation of vascular endothelial growth factor receptors (VEGFRs) by IAI and DME pathophysiological processes. We also undertook an enrichment analysis to explore the processes modulated by IAI, visualized the effectors' predicted protein activity, and specifically evaluated the role of VEGFR1 pathway inhibition on DME treatment. The models simulated the potential pathophysiology of DME and the likely IAI's MoA by inhibiting VEGFR1 and VEGFR2 signaling. The action of IAI through both signaling pathways modulated the identified pathophysiological processes associated with DME, with the strongest effects in angiogenesis, blood-retinal barrier alteration and permeability, and inflammation. VEGFR1 inhibition was essential to modulate inflammatory protein effectors. Given the role of VEGFR1 signaling on the modulation of inflammatory-related pathways, IAI may offer therapeutic advantages for DME through sustained VEGFR1 pathway inhibition.
Collapse
Affiliation(s)
- Morgane Blanot
- Anaxomics Biotech S.L., 08007 Barcelona, Spain; (M.B.); (E.R.); (C.S.-V.); (L.A.)
| | - Ricardo Pedro Casaroli-Marano
- Department of Surgery (FMCS), Universitat de Barcelona, 08007 Barcelona, Spain
- Hospital Clínic de Barcelona (IDIBAPS), Universitat de Barcelona, 08007 Barcelona, Spain
| | | | - Thaïs Sallén
- Bayer Hispania S.L., 08970 Sant Joan Despí, Spain; (J.M.-M.); (T.S.)
| | - Esther Ramírez
- Anaxomics Biotech S.L., 08007 Barcelona, Spain; (M.B.); (E.R.); (C.S.-V.); (L.A.)
| | - Cristina Segú-Vergés
- Anaxomics Biotech S.L., 08007 Barcelona, Spain; (M.B.); (E.R.); (C.S.-V.); (L.A.)
- Research Programme on Biomedical Informatics (GRIB), Departament de Ciències Experimentals i de la Salut, Universitat Pompeu Fabra, 08002 Barcelona, Spain
| | - Laura Artigas
- Anaxomics Biotech S.L., 08007 Barcelona, Spain; (M.B.); (E.R.); (C.S.-V.); (L.A.)
| |
Collapse
|
14
|
Gujarati NA, Chow AK, Mallipattu SK. Central role of podocytes in mediating cellular cross talk in glomerular health and disease. Am J Physiol Renal Physiol 2024; 326:F313-F325. [PMID: 38205544 PMCID: PMC11207540 DOI: 10.1152/ajprenal.00328.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
Podocytes are highly specialized epithelial cells that surround the capillaries of the glomeruli in the kidney. Together with the glomerular endothelial cells, these postmitotic cells are responsible for regulating filtrate from the circulating blood with their organized network of interdigitating foot processes that wrap around the glomerular basement membrane. Although podocyte injury and subsequent loss is the hallmark of many glomerular diseases, recent evidence suggests that the cell-cell communication between podocytes and other glomerular and nonglomerular cells is critical for the development and progression of kidney disease. In this review, we highlight these key cellular pathways of communication and how they might be a potential target for therapy in glomerular disease. We also postulate that podocytes might serve as a central hub for communication in the kidney under basal conditions and in response to cellular stress, which may have implications for the development and progression of glomerular diseases.
Collapse
Affiliation(s)
- Nehaben A Gujarati
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Andrew K Chow
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
| | - Sandeep K Mallipattu
- Division of Nephrology, Department of Medicine, Stony Brook University, Stony Brook, New York, United States
- Renal Section, Northport Veterans Affairs Medical Center, Northport, New York, United States
| |
Collapse
|
15
|
Santorsola M, Capuozzo M, Nasti G, Sabbatino F, Di Mauro A, Di Mauro G, Vanni G, Maiolino P, Correra M, Granata V, Gualillo O, Berretta M, Ottaiano A. Exploring the Spectrum of VEGF Inhibitors' Toxicities from Systemic to Intra-Vitreal Usage in Medical Practice. Cancers (Basel) 2024; 16:350. [PMID: 38254839 PMCID: PMC10813960 DOI: 10.3390/cancers16020350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 01/11/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
The use of Vascular Endothelial Growth Factor inhibitors (VEGFi) has become prevalent in the field of medicine, given the high incidence of various pathological conditions necessitating VEGF inhibition within the general population. These conditions encompass a range of advanced neoplasms, such as colorectal cancer, non-small cell lung cancer, renal cancer, ovarian cancer, and others, along with ocular diseases. The utilization of VEGFi is not without potential risks and adverse effects, requiring healthcare providers to be well-prepared for identification and management. VEGFi can be broadly categorized into two groups: antibodies or chimeric proteins that specifically target VEGF (bevacizumab, ramucirumab, aflibercept, ranibizumab, and brolucizumab) and non-selective and selective small molecules (sunitinib, sorafenib, cabozantinib, lenvatinib, regorafenib, etc.) designed to impede intracellular signaling of the VEGF receptor (RTKi, receptor tyrosine kinase inhibitors). The presentation and mechanisms of adverse effects resulting from VEGFi depend primarily on this distinction and the route of drug administration (systemic or intra-vitreal). This review provides a thorough examination of the causes, recognition, management, and preventive strategies for VEGFi toxicities with the goal of offering support to oncologists in both clinical practice and the design of clinical trials.
Collapse
Affiliation(s)
- Mariachiara Santorsola
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| | | | - Guglielmo Nasti
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| | - Francesco Sabbatino
- Oncology Unit, Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Salerno, Italy;
| | - Annabella Di Mauro
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| | - Giordana Di Mauro
- Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy;
| | - Gianluca Vanni
- Breast Unit, Department of Surgical Science, PTV Policlinico Tor Vergata University, 00133 Rome, Italy;
| | - Piera Maiolino
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| | - Marco Correra
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| | - Vincenza Granata
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude), NEIRID Laboratory (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), IDIS (Instituto de Investigación Sanitaria de Santiago), Research Laboratory 9, Santiago University Clinical Hospital, 15706 Santiago de Compostela, Spain;
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Alessandro Ottaiano
- Istituto Nazionale Tumori di Napoli, IRCCS “G. Pascale”, via M. Semmola, 80131 Naples, Italy; (M.S.); (G.N.); (A.D.M.); (P.M.); (M.C.); (V.G.)
| |
Collapse
|
16
|
Ya J, Pellumbaj J, Hashmat A, Bayraktutan U. The Role of Stem Cells as Therapeutics for Ischaemic Stroke. Cells 2024; 13:112. [PMID: 38247804 PMCID: PMC10814781 DOI: 10.3390/cells13020112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 01/23/2024] Open
Abstract
Stroke remains one of the leading causes of death and disability worldwide. Current reperfusion treatments for ischaemic stroke are limited due to their narrow therapeutic window in rescuing ischaemic penumbra. Stem cell therapy offers a promising alternative. As a regenerative medicine, stem cells offer a wider range of treatment strategies, including long-term intervention for chronic patients, through the reparation and replacement of injured cells via mechanisms of differentiation and proliferation. The purpose of this review is to evaluate the therapeutic role of stem cells for ischaemic stroke. This paper discusses the pathology during acute, subacute, and chronic phases of cerebral ischaemic injury, highlights the mechanisms involved in mesenchymal, endothelial, haematopoietic, and neural stem cell-mediated cerebrovascular regeneration, and evaluates the pre-clinical and clinical data concerning the safety and efficacy of stem cell-based treatments. The treatment of stroke patients with different types of stem cells appears to be safe and efficacious even at relatively higher concentrations irrespective of the route and timing of administration. The priming or pre-conditioning of cells prior to administration appears to help augment their therapeutic impact. However, larger patient cohorts and later-phase trials are required to consolidate these findings.
Collapse
Affiliation(s)
| | | | | | - Ulvi Bayraktutan
- Academic Unit of Mental Health and Clinical Neurosciences, Queens Medical Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
17
|
Guo GX, Wu KY, Zhang XY, Lai FX, Tsim KWK, Qin QW, Hu WH. The extract of Curcumae Longae Rhizoma suppresses angiogenesis via VEGF-induced PI3K/Akt-eNOS-NO pathway. JOURNAL OF ETHNOPHARMACOLOGY 2023; 308:116299. [PMID: 36842721 DOI: 10.1016/j.jep.2023.116299] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/10/2023] [Accepted: 02/16/2023] [Indexed: 06/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Curcumae Longae Rhizoma (CLR) is a safe natural herbal medicine, and which has been widely used for centuries as functional food and health products, but its effects on angiogenesis and related underlying mechanism remain unclear. AIM OF THE STUDY The abnormal angiogenesis is closely related with various diseases, and therefore the precise control of angiogenesis is of great importance. The well-known angiogenic factor, vascular endothelial growth factor (VEGF), mediates angiogenesis and induces multiple signalling pathways via binding to VEGF receptor (VEGFR). The attenuation of VEGF-triggered angiogenic-related signalling pathways may relieve various diseases through suppression of angiogenesis. Here, we aimed to elucidate that CLR extract could exert striking anti-angiogenic activities both in vitro and in vivo. MATERIALS AND METHODS The viability of human umbilical vascular endothelial cell (HUVEC) was examined by LDH and MTT assays. Migrative and invasive ability of the endothelial cells were independently evaluated by wound healing and transwell assays. The activities of CLR extract on in vitro angiogenesis was tested by tube formation assay. In vivo vascularization was determined by using zebrafish embryo model in the present of CLR extract. Western blotting was applied to determine the phosphorylated levels of VEGFR2, PI3K, AKT and eNOS. Besides, the levels of nitric oxide (NO) and reactive oxygen species (ROS) were separately evaluated by Griess assay and 2'7'-dichlorofluorescein diacetate reaction. In addition, the cell migrative ability of cancer cell was estimated by using cultured human colon carcinoma cells (HT-29 cell line), and immunofluorescence assay was applied to evaluate the effect of CLR extract on nuclear translocation of NF-κB p65 subunit in the VEGF-treated HT-29 cultures. RESULTS CLR extract significantly suppressed a series of VEGF-mediated angiogenic responses, including endothelial cell proliferation, migration, invasion, and tube formation. Moreover, CLR extract reduced in vivo sub-intestinal vessel formation in zebrafish embryo model. Mechanistically, the extract of CLR attenuated the VEGF-triggered signalling, as demonstrated by decreased level of phosphorylated VEGFR2 and subsequently inactivated its downstream regulators, e.g. phospho-PI3K, phospho-AKT and phospho-eNOS. The production of NO and formation of ROS were markedly inhibited in HUVECs. Furthermore, CLR extract suppressed cell migration and NF-κB translocation in cultured HT-29 cells. CONCLUSIONS These preclinical findings demonstrate that the extract of CLR remarkably attenuates angiogenesis and which has great potential as a natural drug candidate with excellent anti-angiogenic activity.
Collapse
Affiliation(s)
- Guo-Xia Guo
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China.
| | - Ke-Yue Wu
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China.
| | - Xiao-Yong Zhang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China.
| | - Fu-Xiang Lai
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China.
| | - Karl Wah-Keung Tsim
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China; Division of Life Science and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China.
| | - Qi-Wei Qin
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China.
| | - Wei-Hui Hu
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, China; University Joint Laboratory of Guangdong Province, Hong Kong and Macao Region on Marine Bioresource Conservation and Exploitation, Guangzhou, China.
| |
Collapse
|
18
|
Zaied AA, Ushio‐Fukai M, Fukai T, Kovacs‐Kasa A, Alhusban S, Sudhahar V, Ganta VC, Annex BH. Pentose Pathway Activation Is Superior to Increased Glycolysis for Therapeutic Angiogenesis in Peripheral Arterial Disease. J Am Heart Assoc 2023; 12:e027986. [PMID: 36974760 PMCID: PMC10122893 DOI: 10.1161/jaha.122.027986] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/05/2022] [Indexed: 03/29/2023]
Abstract
Background In endothelial cells (ECs), glycolysis, regulated by PFKFB3 (6-phosphofructo-2-kinase/fructose-2,6-biphosphatase, isoform-3), is the major metabolic pathway for ATP generation. In preclinical peripheral artery disease models, VEGF165a (vascular endothelial growth factor165a) and microRNA-93 both promote angiogenesis. Methods and Results Mice following hind-limb ischemia (HLI) and ECs with, and without, hypoxia and serum starvation were examined with, and without, microRNA-93 and VEGF165a. Post-HLI perfusion recovery was monitored. EC metabolism was studied using seahorse assay, and the expression and activity of major metabolism genes were assessed. Reactive oxygen species levels and EC permeability were evaluated. C57Bl/6J mice generated a robust angiogenic response to HLI, with ECs from ischemic versus nonischemic muscle demonstrating no increase in glycolysis. Balb/CJ mice generated a poor angiogenic response post-HLI; ischemic versus nonischemic ECs demonstrated significant increase in glycolysis. MicroRNA-93-treated Balb/CJ mice post-HLI showed better perfusion recovery, with ischemic versus nonischemic ECs showing no increase in glycolysis. VEGF165a-treated Balb/CJ mice post-HLI showed no improvement in perfusion recovery with ischemic versus nonischemic ECs showing significant increase in glycolysis. ECs under hypoxia and serum starvation upregulated PFKFB3. In ECs under hypoxia and serum starvation, VEGF165a versus control significantly upregulated PFKFB3 and glycolysis, whereas miR-93 versus control demonstrated no increase in PFKFB3 or glycolysis. MicroRNA-93 versus VEGF165a upregulated glucose-6-phosphate dehydrogenase expression and activity, activating the pentose phosphate pathway. MicroRNA-93 versus control increased reduced nicotinamide adenine dinucleotide phosphate and virtually eliminated the increase in reactive oxygen species. In ECs under hypoxia and serum starvation, VEGF165a significantly increased and miR-93 decreased EC permeability. Conclusions In peripheral artery disease, activation of the pentose phosphate pathway to promote angiogenesis may offer potential therapeutic advantages.
Collapse
Affiliation(s)
- Abdelrahman A. Zaied
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
- Department of MedicineMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Masuko Ushio‐Fukai
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Tohru Fukai
- Departments of Pharmacology and ToxicologyMedical College of Georgia at Augusta UniversityAugustaGAUSA
- Charlie Norwood Veterans Affairs Medical CenterAugustaGAUSA
| | - Anita Kovacs‐Kasa
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Suhib Alhusban
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Varadarajan Sudhahar
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Vijay C. Ganta
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Brian H. Annex
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
- Department of MedicineMedical College of Georgia at Augusta UniversityAugustaGAUSA
| |
Collapse
|
19
|
Gallo G, Lanza O, Savoia C. New Insight in Cardiorenal Syndrome: From Biomarkers to Therapy. Int J Mol Sci 2023; 24:5089. [PMID: 36982164 PMCID: PMC10049666 DOI: 10.3390/ijms24065089] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/09/2023] Open
Abstract
Cardiorenal syndrome consists in the coexistence of acute or chronic dysfunction of heart and kidneys resulting in a cascade of feedback mechanisms and causing damage to both organs associated with high morbidity and mortality. In the last few years, different biomarkers have been investigated with the aim to achieve an early and accurate diagnosis of cardiorenal syndrome, to provide a prognostic role and to guide the development of targeted pharmacological and non-pharmacological therapies. In such a context, sodium-glucose cotransporter 2 (SGLT2) inhibitors, recommended as the first-line choice in the management of heart failure, might represent a promising strategy in the management of cardiorenal syndrome due to their efficacy in reducing both cardiac and renal outcomes. In this review, we will discuss the current knowledge on the pathophysiology of cardiorenal syndrome in adults, as well as the utility of biomarkers in cardiac and kidney dysfunction and potential insights into novel therapeutics.
Collapse
Affiliation(s)
| | | | - Carmine Savoia
- Clinical and Molecular Medicine Department, Faculty of Medicine and Psychology, Sant’Andrea Hospital, Sapienza University of Rome, 00189 Rome, Italy
| |
Collapse
|
20
|
Mohandes S, Doke T, Hu H, Mukhi D, Dhillon P, Susztak K. Molecular pathways that drive diabetic kidney disease. J Clin Invest 2023; 133:165654. [PMID: 36787250 PMCID: PMC9927939 DOI: 10.1172/jci165654] [Citation(s) in RCA: 140] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
Kidney disease is a major driver of mortality among patients with diabetes and diabetic kidney disease (DKD) is responsible for close to half of all chronic kidney disease cases. DKD usually develops in a genetically susceptible individual as a result of poor metabolic (glycemic) control. Molecular and genetic studies indicate the key role of podocytes and endothelial cells in driving albuminuria and early kidney disease in diabetes. Proximal tubule changes show a strong association with the glomerular filtration rate. Hyperglycemia represents a key cellular stress in the kidney by altering cellular metabolism in endothelial cells and podocytes and by imposing an excess workload requiring energy and oxygen for proximal tubule cells. Changes in metabolism induce early adaptive cellular hypertrophy and reorganization of the actin cytoskeleton. Later, mitochondrial defects contribute to increased oxidative stress and activation of inflammatory pathways, causing progressive kidney function decline and fibrosis. Blockade of the renin-angiotensin system or the sodium-glucose cotransporter is associated with cellular protection and slowing kidney function decline. Newly identified molecular pathways could provide the basis for the development of much-needed novel therapeutics.
Collapse
Affiliation(s)
- Samer Mohandes
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tomohito Doke
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hailong Hu
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Dhanunjay Mukhi
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Poonam Dhillon
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Katalin Susztak
- Renal, Electrolyte, and Hypertension Division, Department of Medicine;,Institute for Diabetes, Obesity, and Metabolism;,Department of Genetics; and,Kidney Innovation Center; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
21
|
Ribatti D, Ligresti G, Nicosia RF. Kidney endothelial cell heterogeneity, angiocrine activity and paracrine regulatory mechanisms. Vascul Pharmacol 2023; 148:107139. [PMID: 36539108 PMCID: PMC10828957 DOI: 10.1016/j.vph.2022.107139] [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] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/07/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022]
Abstract
The blood microvascular endothelium consists of a heterogeneous population of cells with regionally distinct morphologies and transcriptional signatures in different tissues and organs. In addition to providing an anti-thrombogenic surface for blood flow, endothelial cells perform a multitude of additional regulatory tasks involving organogenesis, metabolism, angiogenesis, inflammation, repair and organ homeostasis. To communicate with surrounding cells and accomplish their many functions, endothelial cells secrete angiocrine factors including growth factors, chemokines, cytokines, extracellular matrix components, and proteolytic enzymes. Nonendothelial parenchymal and stromal cells in turn regulate endothelial growth, differentiation and survival during embryonal development and in the adult by paracrine mechanisms. Driven by advances in molecular biology, animal genetics, single cell transcriptomics and microscopic imaging, knowledge of organotypic vasculatures has expanded rapidly in recent years. The kidney vasculature, in particular, has been the focus of intensive investigation and represents a primary example of how endothelial heterogeneity and crosstalk with nonendothelial cells contribute to the development and function of a vital organ. In this paper, we review the morphology, function, and development of the kidney vasculature, with an emphasis on blood microvascular endothelial heterogeneity, and provide examples of endothelial and nonendothelial-derived factors that are critically involved in kidney development, growth, response to injury, and homeostasis.
Collapse
Affiliation(s)
- Domenico Ribatti
- Dipartimento di Scienze Mediche di Base, Neuroscienze e Organi di Senso (SMBNOS), Universita' degli Studi Aldo Moro, Policlinico, Piazza G. Cesare, 11, - Bari, Italy.
| | - Giovanni Ligresti
- Department of Medicine, Boston University School of Medicine, 72 East Concord Street, Boston, MA 02118, United States of America
| | - Roberto F Nicosia
- Department of Laboratory Medicine and Pathology, University of Washington, Box 356100, 1959 NE Pacific St, Seattle, WA 98195, United States of America
| |
Collapse
|
22
|
Basu D, Pal R, Sarkar M, Barma S, Halder S, Roy H, Nandi S, Samadder A. To Investigate Growth Factor Receptor Targets and Generate Cancer Targeting Inhibitors. Curr Top Med Chem 2023; 23:2877-2972. [PMID: 38164722 DOI: 10.2174/0115680266261150231110053650] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/20/2023] [Accepted: 10/02/2023] [Indexed: 01/03/2024]
Abstract
Receptor tyrosine kinase (RTK) regulates multiple pathways, including Mitogenactivated protein kinases (MAPKs), PI3/AKT, JAK/STAT pathway, etc. which has a significant role in the progression and metastasis of tumor. As RTK activation regulates numerous essential bodily processes, including cell proliferation and division, RTK dysregulation has been identified in many types of cancers. Targeting RTK is a significant challenge in cancer due to the abnormal upregulation and downregulation of RTK receptors subfamily EGFR, FGFR, PDGFR, VEGFR, and HGFR in the progression of cancer, which is governed by multiple RTK receptor signalling pathways and impacts treatment response and disease progression. In this review, an extensive focus has been carried out on the normal and abnormal signalling pathways of EGFR, FGFR, PDGFR, VEGFR, and HGFR and their association with cancer initiation and progression. These are explored as potential therapeutic cancer targets and therefore, the inhibitors were evaluated alone and merged with additional therapies in clinical trials aimed at combating global cancer.
Collapse
Affiliation(s)
- Debroop Basu
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Riya Pal
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, IndiaIndia
| | - Maitrayee Sarkar
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Soubhik Barma
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Sumit Halder
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| | - Harekrishna Roy
- Nirmala College of Pharmacy, Vijayawada, Guntur, Andhra Pradesh, India
| | - Sisir Nandi
- Global Institute of Pharmaceutical Education and Research (Affiliated to Uttarakhand Technical University), Kashipur, 244713, India
| | - Asmita Samadder
- Cell and Developmental Biology Special, Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
- Cytogenetics and Molecular Biology Lab., Department of Zoology, University of Kalyani, Kalyani, Nadia, 741235, India
| |
Collapse
|
23
|
Dela Justina V, Dos Passos Júnior RR, Lima VV, Giachini FR. Evidence of Nitric Oxide Impairment During Hypertensive Pregnancies. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:99-125. [PMID: 37466771 DOI: 10.1007/978-3-031-32554-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Hypertensive disorders of pregnancy complicate up to 10% of pregnancies worldwide, and they can be classified into (1) gestational hypertension, (2) preeclampsia, (3) chronic hypertension and (4) chronic hypertension with preeclampsia. Nitric oxide (NO) plays an essential role in the haemodynamic adaptations observed during pregnancy. It has been shown that the nitric oxide pathway's dysfunction during pregnancy is associated with placental- and vascular-related diseases such as hypertensive disorders of pregnancy. This review aims to present a brief definition of hypertensive disorders of pregnancy and physiological maternal cardiovascular adaptations during pregnancy. We also detail how NO signalling is altered in the (a) systemic vasculature, (b) uterine artery/spiral arteries, (c) implantation and (d) placenta of hypertensive disorders during pregnancy. We conclude by summarizing the anti-hypertensive therapy of hypertensive disorders of pregnancy as a specific management strategy.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
| | - Rinaldo Rodrigues Dos Passos Júnior
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
- Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra do Garcas, Brazil
| | - Victor Vitorino Lima
- Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra do Garcas, Brazil
| | - Fernanda Regina Giachini
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, Brazil
- Institute of Health Sciences and Health, Universidad Federal De Mato Grosso, Barra do Garcas, Brazil
| |
Collapse
|
24
|
Sun H, Morihara R, Feng T, Bian Z, Yu H, Hu X, Hu X, Bian Y, Sasaki R, Fukui Y, Takemoto M, Yunoki T, Nakano Y, Abe K, Yamashita T. Human Cord Blood-Endothelial Progenitor Cells Alleviate Intimal Hyperplasia of Arterial Damage in a Rat Stroke Model. Cell Transplant 2023; 32:9636897231193069. [PMID: 37615293 PMCID: PMC10467372 DOI: 10.1177/09636897231193069] [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: 03/26/2023] [Revised: 06/28/2023] [Accepted: 07/18/2023] [Indexed: 08/25/2023] Open
Abstract
Human cord blood-endothelial progenitor cells (hCB-EPCs) isolated from the human umbilical cord can be used to repair damaged arteries. In this study, we used an animal model with pathological changes that mimics artery wall damage caused by stent retrievers in humans. We injected hCB-EPCs to investigate their effect on endothelial hyperplasia and dysfunction during intimal repair. Four groups were established based on the length of reperfusion (3 and 28 days), as well as the presence or absence of hCB-EPC therapy. Damage to the internal carotid artery was evaluated by hematoxylin-eosin and immunohistochemical staining. Stroke volume was not significantly different between non-EPC and EPC groups although EPC treatment alleviated intimal hyperplasia 28 days after intimal damage. Vascular endothelial growth factor (VEGF) and eNOS expression were significantly higher in the EPC-treated group than in the non-EPC group 3 days after intimal damage. In addition, MMP9 and 4HNE expression in the EPC-treated group was significantly lower than in the non-EPC group. Ultimately, this study found that venous transplantation of hCB-EPCs could inhibit neointimal hyperplasia, alleviate endothelial dysfunction, suppress intimal inflammation, and reduce oxidative stress during healing of intimal damage.
Collapse
Affiliation(s)
- Hongming Sun
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Tian Feng
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Zhihong Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Haibo Yu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Xiao Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Xinran Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yuting Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ryo Sasaki
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yusuke Fukui
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Mami Takemoto
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Taijun Yunoki
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Yumiko Nakano
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Koji Abe
- National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
25
|
Yin T, Chen S, Zeng G, Yuan W, Lu Y, Zhang Y, Huang Q, Xiong X, Xu B, Huang Q. Angiogenesis-Browning Interplay Mediated by Asprosin-Knockout Contributes to Weight Loss in Mice with Obesity. Int J Mol Sci 2022; 23:16166. [PMID: 36555807 PMCID: PMC9783228 DOI: 10.3390/ijms232416166] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 12/09/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Asprosin (ASP) is a recently identified adipokine secreted by white adipose tissue (WAT). It plays important roles in the maintenance of glucose homeostasis in the fasting state and in the occurrence and development of obesity. However, there is no report on whether and how ASP would inhibit angiogenesis and fat browning in the mouse adipose microenvironment. Therefore, the study sought to investigate the effects of ASP-knockout on angiogenesis and fat browning, and to identify the interaction between them in the ASP-knockout mouse adipose microenvironment. In the experiments in vivo, the ASP-knockout alleviated the obesity induced by a high fat diet (HFD) and increased the expressions of the browning-related proteins including uncoupling protein 1 (UCP1), PRD1-BF-1-RIZ1 homologus domain-containing protein-16 (PRDM16) and PPAR gamma coactivator 1 (PGC1-α) and the endothelial cell marker (CD31). In the experiments in vitro, treatment with the conditional medium (CM) from ASP-knockout adipocytes (ASP-/--CM) significantly promoted the proliferation, migration and angiogenesis of vascular endothelial cells, and increased the expressions of vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor 2 (VEGFR2) and phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/endothelial nitric oxide synthase (eNOS) pathway proteins. In addition, the treatment with CM from endothelial cells (EC-CM) markedly reduced the accumulation of lipid droplets and increased the expressions of the browning-related proteins and the mitochondrial contents. Moreover, the treatment with EC-CM significantly improved the energy metabolism in 3T3-L1 adipocytes. These results highlight that ASP-knockout can promote the browning and angiogenesis of WAT, and the fat browning and angiogenesis can interact in the mouse adipose microenvironment, which contributes to weight loss in the mice with obesity.
Collapse
Affiliation(s)
- Tingting Yin
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Sheng Chen
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Guohua Zeng
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Wanwan Yuan
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Yanli Lu
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Yanan Zhang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Qianqian Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Xiaowei Xiong
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Baohua Xu
- Jiangxi Province Key Laboratory of Laboratory Animal, Nanchang 330006, China
| | - Qiren Huang
- Key Provincial Laboratory of Basic Pharmacology, Nanchang University, Nanchang 330006, China
- Department of Pharmacology, School of Pharmacy, Nanchang University, Nanchang 330006, China
| |
Collapse
|
26
|
Miao C, Zhu X, Wei X, Long M, Jiang L, Li C, Jin D, Du Y. Pro- and anti-fibrotic effects of vascular endothelial growth factor in chronic kidney diseases. Ren Fail 2022; 44:881-892. [PMID: 35618410 PMCID: PMC9154791 DOI: 10.1080/0886022x.2022.2079528] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Renal fibrosis is the inevitable common end-point of all progressive chronic kidney diseases. The underlying mechanisms of renal fibrosis are complex, and currently there is no effective therapy against renal fibrosis. Renal microvascular rarefaction contributes to the progression of renal fibrosis; however, an imbalance between proangiogenic and antiangiogenic factors leads to the loss of renal microvasculature. Vascular endothelial growth factor (VEGF) is the most important pro-angiogenic factor. Recent studies have unraveled the involvement of VEGF in the regulation of renal microvascular rarefaction and fibrosis via various mechanisms; however, it is not clear whether it has anti-fibrotic or pro-fibrotic effect. This paper reviews the available evidence pertaining to the function of VEGF in the fibrotic process and explores the associated underlying mechanisms. Our synthesis will help identify the future research priorities for developing specialized treatments for alleviating or preventing renal fibrosis. Abbreviation: VEGF: vascular endothelial growth factor; CKD: chronic kidney disease; ESKD: end-stage kidney disease; ER: endoplasmic reticulum; VEGFR: vascular endothelial growth factor receptor; AKI: acute kidney injury; EMT: epithelial-to-mesenchymal transition; HIF: hypoxia-inducible factor; α-SMA: α smooth muscle actin; UUO: unilateral ureteral obstruction; TGF-β: transforming growth factor-β; PMT: pericyte-myofibroblast transition; NO: nitric oxide; NOS: nitric oxide synthase; nNOS: neuronal nitric oxide synthase; iNOS: inducible nitric oxide synthase; eNOS: endothelial nitric oxide synthase; sGC: soluble guanylate cyclase; PKG: soluble guanylate cyclase dependent protein kinases; UP R: unfolded protein response
Collapse
Affiliation(s)
- Changxiu Miao
- Department of Nephrology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Xiaoyu Zhu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Xuejiao Wei
- Department of Nephrology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Mengtuan Long
- Department of Nephrology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Lili Jiang
- Physical Examination Center, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Chenhao Li
- Department of Nephrology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Die Jin
- Department of Nephrology, The First Hospital of Jilin University, Changchun, People's Republic of China
| | - Yujun Du
- Department of Nephrology, The First Hospital of Jilin University, Changchun, People's Republic of China
| |
Collapse
|
27
|
Veron D, Aggarwal PK, Li Q, Moeckel G, Kashgarian M, Tufro A. Podocyte VEGF-A Knockdown Induces Diffuse Glomerulosclerosis in Diabetic and in eNOS Knockout Mice. Front Pharmacol 2022; 12:788886. [PMID: 35280251 PMCID: PMC8906751 DOI: 10.3389/fphar.2021.788886] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/13/2021] [Indexed: 01/06/2023] Open
Abstract
Vascular endothelial growth factor-a (VEGF-A) and nitric oxide (NO) are essential for glomerular filtration barrier homeostasis, and are dysregulated in diabetic kidney disease (DKD). While NO availability is consistently low in diabetes, both high and low VEGF-A have been reported in patients with DKD. Here we examined the effect of inducible podocyte VEGF-A knockdown (VEGFKD) in diabetic mice and in endothelial nitric oxide synthase knockout mice (eNOS−/−). Diabetes was induced with streptozotocin using the Animal Models of Diabetic Complications Consortium (AMDCC) protocol. Induction of podocyte VEGFKD led to diffuse glomerulosclerosis, foot process effacement, and GBM thickening in both diabetic mice with intact eNOS and in non-diabetic eNOS−/−:VEGFKD mice. VEGFKD diabetic mice developed mild proteinuria and maintained normal glomerular filtration rate (GFR), associated with extremely high NO and thiol urinary excretion. In eNOS−/−:VEGFKD (+dox) mice severe diffuse glomerulosclerosis was associated with microaneurisms, arteriolar hyalinosis, massive proteinuria, and renal failure. Collectively, data indicate that combined podocyte VEGF-A and eNOS deficiency result in diffuse glomerulosclerosis in mice; compensatory NO and thiol generation prevents severe proteinuria and GFR loss in VEGFKD diabetic mice with intact eNOS, whereas VEGFKD induction in eNOS−/−:VEGFKD mice causes massive proteinuria and renal failure mimicking DKD in the absence of diabetes. Mechanistically, we identify VEGFKD-induced abnormal S-nitrosylation of specific proteins, including β3-integrin, laminin, and S-nitrosoglutathione reductase (GSNOR), as targetable molecular mechanisms involved in the development of advanced diffuse glomerulosclerosis and renal failure.
Collapse
Affiliation(s)
- Delma Veron
- Department of Pediatrics, Yale University School of Medicine, Malvern, PA, United States
| | - Pardeep K Aggarwal
- Department of Pediatrics, Yale University School of Medicine, Malvern, PA, United States
| | - Qi Li
- Department of Pediatrics, Yale University School of Medicine, Malvern, PA, United States.,Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Gilbert Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Michael Kashgarian
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Alda Tufro
- Department of Pediatrics, Yale University School of Medicine, Malvern, PA, United States.,Department of Cell and Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
28
|
Bevacizumab Increases Endothelin-1 Production via Forkhead Box Protein O1 in Human Glomerular Microvascular Endothelial Cells In Vitro. Int J Nephrol 2021; 2021:8381115. [PMID: 34912580 PMCID: PMC8668358 DOI: 10.1155/2021/8381115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/15/2021] [Indexed: 11/17/2022] Open
Abstract
Molecular mechanisms underlying the nephrotoxicity associated with bevacizumab are unclear. Endothelin-1 (ET-1) is involved in podocyte injury and proteinuria, and its level increases in most cases of kidney disorders. Forkhead box protein O1 (FoxO1), a transcription factor, is a major determinant of ET-1 promoter activation and is regulated by protein kinase B (Akt) phosphorylation-dependent nuclear exclusion. We evaluated the effect of bevacizumab on ET-1 production in human glomerular microvascular endothelial cells (hGECs). We analyzed the changes in the mRNA and protein levels of ET-1 in hGECs treated with bevacizumab using real-time reverse transcription-polymerase chain reaction and enzyme-linked immunosorbent assay. Changes in the protein levels and phosphorylation status of Akt and FoxO1 in hGECs treated with bevacizumab were analyzed by western blotting. After cell lysis, FoxO1 protein was isolated from the cytoplasmic and nuclear fractions. We also investigated the effects of AS1842856 (a FoxO1 inhibitor) on bevacizumab-induced ET-1 production. Bevacizumab significantly and dose-dependently increased the mRNA and protein levels of ET-1 in hGECs (p < 0.05). Bevacizumab treatment also led to a decrease in phosphorylated Akt protein levels. Inhibition of Akt activity by LY294002 promoted ET-1 production. Bevacizumab also induced an increase in FoxO1 protein levels in the nucleus. Inhibition of FoxO1 activity by AS1842856 resulted in decreased ET-1 levels in bevacizumab-treated hGECs. ET-1 axis activation, Akt inactivation, and FoxO1 nuclear localization are the molecular mechanisms underlying bevacizumab-induced nephrotoxicity. Therefore, inhibition of renal ET-1 production could be a promising approach to protect against or treat bevacizumab-induced nephrotoxicity.
Collapse
|
29
|
Repurposing Riociguat to Target a Novel Paracrine Nitric Oxide-TRPC6 Pathway to Prevent Podocyte Injury. Int J Mol Sci 2021; 22:ijms222212485. [PMID: 34830371 PMCID: PMC8621407 DOI: 10.3390/ijms222212485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023] Open
Abstract
Increased expression and activity of the Ca2+ channel transient receptor potential channel 6 (TRPC6) is associated with focal segmental glomerulosclerosis, but therapeutic strategies to target TRPC6 are currently lacking. Nitric oxide (NO) is crucial for normal glomerular function and plays a protective role in preventing glomerular diseases. We investigated if NO prevents podocyte injury by inhibiting injurious TRPC6-mediated signaling in a soluble guanylate cyclase (sGC)-dependent manner and studied the therapeutic potential of the sGC stimulator Riociguat. Experiments were performed using human glomerular endothelial cells and podocytes. Podocyte injury was induced by Adriamycin incubation for 24 h, with or without the NO-donor S-Nitroso-N-acetyl-DL-penicillamine (SNAP), the sGC stimulator Riociguat or the TRPC6 inhibitor Larixyl Acetate (LA). NO and Riociguat stimulated cGMP synthesis in podocytes, decreased Adriamycin-induced TRPC6 expression, inhibited the Adriamycin-induced TRPC6-mediated Ca2+ influx and reduced podocyte injury. The protective effects of Riociguat and NO were blocked when sGC activity was inhibited with 1H-[1,2,4]Oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) or when TRPC6 activity was inhibited by LA. Our data demonstrate a glomerular (e)NOS-NO-sGC-cGMP-TRPC6 pathway that prevents podocyte injury, which can be translated to future clinical use by, e.g., repurposing the market-approved drug Riociguat.
Collapse
|
30
|
Opichka MA, Rappelt MW, Gutterman DD, Grobe JL, McIntosh JJ. Vascular Dysfunction in Preeclampsia. Cells 2021; 10:3055. [PMID: 34831277 PMCID: PMC8616535 DOI: 10.3390/cells10113055] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/01/2021] [Accepted: 11/04/2021] [Indexed: 01/22/2023] Open
Abstract
Preeclampsia is a life-threatening pregnancy-associated cardiovascular disorder characterized by hypertension and proteinuria at 20 weeks of gestation. Though its exact underlying cause is not precisely defined and likely heterogenous, a plethora of research indicates that in some women with preeclampsia, both maternal and placental vascular dysfunction plays a role in the pathogenesis and can persist into the postpartum period. Potential abnormalities include impaired placentation, incomplete spiral artery remodeling, and endothelial damage, which are further propagated by immune factors, mitochondrial stress, and an imbalance of pro- and antiangiogenic substances. While the field has progressed, current gaps in knowledge include detailed initial molecular mechanisms and effective treatment options. Newfound evidence indicates that vasopressin is an early mediator and biomarker of the disorder, and promising future therapeutic avenues include mitigating mitochondrial dysfunction, excess oxidative stress, and the resulting inflammatory state. In this review, we provide a detailed overview of vascular defects present during preeclampsia and connect well-established notions to newer discoveries at the molecular, cellular, and whole-organism levels.
Collapse
Affiliation(s)
- Megan A. Opichka
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.A.O.); (D.D.G.); (J.L.G.)
| | - Matthew W. Rappelt
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
| | - David D. Gutterman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.A.O.); (D.D.G.); (J.L.G.)
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Justin L. Grobe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.A.O.); (D.D.G.); (J.L.G.)
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Comprehensive Rodent Metabolic Phenotyping Core, Medical College of Wisconsin, Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Jennifer J. McIntosh
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; (M.A.O.); (D.D.G.); (J.L.G.)
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA;
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
31
|
Ricciardi CA, Gnudi L. Vascular growth factors as potential new treatment in cardiorenal syndrome in diabetes. Eur J Clin Invest 2021; 51:e13579. [PMID: 33942293 DOI: 10.1111/eci.13579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/11/2021] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Cardiorenal syndrome in diabetes is characterised by alterations of the cardiovascular system paralleled by kidney disease with progressive renal function decline. In diabetes, chronic metabolic and haemodynamic perturbations drive endothelial dysfunction, inflammation, oxidative stress and progressive tissue fibrosis which, in turn, lead to heart and renal anatomo-functional damage. In physiology, vascular growth factors have been implicated in vascular homeostasis; their imbalance, in disease setting such as diabetes, leads to vascular dysfunction and cardiorenal damage. AIMS To define the role of vascular growth factors and angiopoietins in cardiorenal syndrome. MATERIAL AND METHODS We will focus on the two most studied vascular growth factors, vascular endothelial growth factor (VEGF) and angiopoietins (Angpt). The balance and crosstalk between these growth factors are important in organ development and in the maintenance of a healthy vasculature, heart and kidney. The observed alterations in expression/function of these vascular growth factors, as seen in diabetes, are a protective response against external perturbations. RESULTS The chronic insults driving diabetes-mediated cardiorenal damage results in a paradoxical situation, whereby the vascular growth factors imbalance becomes a mechanism of disease. Studies have explored the possibility of modulating the expression/action of vascular growth factors to improve disease outcome. Experimental work has been conducted in animals and has been gradually translated in humans. DISCUSSION Difficulties have been encountered especially when considering the magnitude, timing and duration of interventions targeting a selective vascular growth factor. Targeting VEGF in cardiovascular disease has been challenging, while modulation of the Angpt system seems more promising. CONCLUSION Future studies will establish the translatability of therapies targeting vascular growth factors for heart and kidney disease in patients with diabetes.
Collapse
Affiliation(s)
- Carlo Alberto Ricciardi
- Section Vascular Biology and Inflammation, School of Cardiovascular Medicine & Sciences, British Heart Foundation Centre for Research Excellence, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Luigi Gnudi
- Section Vascular Biology and Inflammation, School of Cardiovascular Medicine & Sciences, British Heart Foundation Centre for Research Excellence, Faculty of Life Sciences & Medicine, King's College London, London, UK
| |
Collapse
|
32
|
Uemura A, Fruttiger M, D'Amore PA, De Falco S, Joussen AM, Sennlaub F, Brunck LR, Johnson KT, Lambrou GN, Rittenhouse KD, Langmann T. VEGFR1 signaling in retinal angiogenesis and microinflammation. Prog Retin Eye Res 2021; 84:100954. [PMID: 33640465 PMCID: PMC8385046 DOI: 10.1016/j.preteyeres.2021.100954] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
Five vascular endothelial growth factor receptor (VEGFR) ligands (VEGF-A, -B, -C, -D, and placental growth factor [PlGF]) constitute the VEGF family. VEGF-A binds VEGF receptors 1 and 2 (VEGFR1/2), whereas VEGF-B and PlGF only bind VEGFR1. Although much research has been conducted on VEGFR2 to elucidate its key role in retinal diseases, recent efforts have shown the importance and involvement of VEGFR1 and its family of ligands in angiogenesis, vascular permeability, and microinflammatory cascades within the retina. Expression of VEGFR1 depends on the microenvironment, is differentially regulated under hypoxic and inflammatory conditions, and it has been detected in retinal and choroidal endothelial cells, pericytes, retinal and choroidal mononuclear phagocytes (including microglia), Müller cells, photoreceptor cells, and the retinal pigment epithelium. Whilst the VEGF-A decoy function of VEGFR1 is well established, consequences of its direct signaling are less clear. VEGFR1 activation can affect vascular permeability and induce macrophage and microglia production of proinflammatory and proangiogenic mediators. However the ability of the VEGFR1 ligands (VEGF-A, PlGF, and VEGF-B) to compete against each other for receptor binding and to heterodimerize complicates our understanding of the relative contribution of VEGFR1 signaling alone toward the pathologic processes seen in diabetic retinopathy, retinal vascular occlusions, retinopathy of prematurity, and age-related macular degeneration. Clinically, anti-VEGF drugs have proven transformational in these pathologies and their impact on modulation of VEGFR1 signaling is still an opportunity-rich field for further research.
Collapse
Affiliation(s)
- Akiyoshi Uemura
- Department of Retinal Vascular Biology, Nagoya City University Graduate School of Medical Sciences, 1 Kawasumi Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan.
| | - Marcus Fruttiger
- UCL Institute of Ophthalmology, University College London, 11-43 Bath Street, London, EC1V 9EL, UK.
| | - Patricia A D'Amore
- Schepens Eye Research Institute of Massachusetts Eye and Ear, 20 Staniford Street, Boston, MA, 02114, USA.
| | - Sandro De Falco
- Angiogenesis Laboratory, Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", Via Pietro Castellino 111, 80131 Naples, Italy; ANBITION S.r.l., Via Manzoni 1, 80123, Naples, Italy.
| | - Antonia M Joussen
- Department of Ophthalmology, Charité-Universitätsmedizin Berlin, Hindenburgdamm 30, 12200 Berlin, and Augustenburger Platz 1, 13353, Berlin, Germany.
| | - Florian Sennlaub
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012, Paris, France.
| | - Lynne R Brunck
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Kristian T Johnson
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - George N Lambrou
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Kay D Rittenhouse
- Bayer Consumer Care AG, Pharmaceuticals, Peter-Merian-Strasse 84, CH-4052 Basel, Switzerland.
| | - Thomas Langmann
- Laboratory for Experimental Immunology of the Eye, Department of Ophthalmology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 9, 50931, Cologne, Germany.
| |
Collapse
|
33
|
Qin-Xiao, Sun YY, Lu ZJ, Zhang TZ, Li SS, Hua T, Suriguga, Chen WL, Ran LL, Yu WZ, Yang F, Burenbatu. Inhibitory effects of safranal on laser-induced choroidal neovascularization and human choroidal microvascular endothelial cells and related pathways analyzed with transcriptome sequencing. Int J Ophthalmol 2021; 14:981-989. [PMID: 34282381 DOI: 10.18240/ijo.2021.07.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/14/2021] [Indexed: 11/23/2022] Open
Abstract
AIM To determine the effects of safranal on choroidal neovascularization (CNV) and oxidative stress damage of human choroidal microvascular endothelial cells (HCVECs) and its possible mechanisms. METHODS Forty-five rats were used as a laser-induced CNV model for testing the efficacy and safety of safranal (0.5 mg/kg·d, intraperitoneally) on CNV. CNV leakage on fluorescein angiography (FA) and CNV thickness on histology was compared. HCVECs were used for a H2O2-induced oxidative stress model to test the effect of safranal in vitro. MTT essay was carried to test the inhibition rate of safranal on cell viability at different concentrations. Tube formation was used to test protective effect of safranal on angiogenesis at different concentrations. mRNA transcriptome sequencing was performed to find the possible signal pathway. The expressions of different molecules and their phosphorylation level were validated by Western blotting. RESULTS On FA, the average CNV leakage area was 0.73±0.49 and 0.31±0.11 mm2 (P=0.012) in the control and safranal-treated group respectively. The average CNV thickness was 127.4±18.75 and 100.6±17.34 µm (P=0.001) in control and safranal-treated group. Under the condition of oxidative stress, cell proliferation was inhibited by safranal and inhibition rates were 7.4%-35.4% at the different concentrations. For tube formation study, the number of new branches was 364 in control group and 35, 42, and 17 in 20, 40, and 80 µg/mL safranal groups respectively (P<0.01). From the KEGG pathway bubble graph, the PI3K-AKT signaling pathway showed a high gene ratio. The protein expression was elevated of insulin receptor substrate (IRS) and the phosphorylation level of PI3K, phosphoinositide-dependent protein kinase 1/2 (PDK1/2), AKT and Bcl-2 associated death promoter (BAD) was also elevated under oxidative stress condition but inhibited by safranal. CONCLUSION Safranal can inhibit CNV both in vivo and in vitro, and the IRS-PI3K-PDK1/2-AKT-BAD signaling pathway is involved in the pathogenesis of CNV.
Collapse
Affiliation(s)
- Qin-Xiao
- Department of Ophthalmology, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao 028007, Inner Mongolia Autonomous Region, China
| | - Yao-Yao Sun
- Department of Ophthalmology, Peking University People's Hospital, Beijing100044, China
| | - Zhan-Jun Lu
- Department of Ophthalmology, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao 028007, Inner Mongolia Autonomous Region, China
| | - Tian-Zi Zhang
- Department of Ophthalmology, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao 028007, Inner Mongolia Autonomous Region, China
| | - Shan-Shan Li
- Department of Ophthalmology, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao 028007, Inner Mongolia Autonomous Region, China
| | - Ting Hua
- College of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Suriguga
- College of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Wen-Lin Chen
- College of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Lin-Lin Ran
- College of Mongolian Medicine, Inner Mongolia University for Nationalities, Tongliao 028000, Inner Mongolia Autonomous Region, China
| | - Wen-Zhen Yu
- Department of Ophthalmology, Peking University People's Hospital, Beijing100044, China
| | - Fei Yang
- Department of Ophthalmology, Peking University International Hospital, Beijing 100026, China
| | - Burenbatu
- Department of Hematology, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao 028007, Inner Mongolia Autonomous Region, China
| |
Collapse
|
34
|
Bubb KJ, Tang O, Gentile C, Moosavi SM, Hansen T, Liu CC, Di Bartolo BA, Figtree GA. FXYD1 Is Protective Against Vascular Dysfunction. Hypertension 2021; 77:2104-2116. [PMID: 33934624 DOI: 10.1161/hypertensionaha.120.16884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Kristen J Bubb
- From the University of Sydney, Kolling Institute of Medical Research, Cardiothoracic and Vascular Health (K.J.B., O.T., C.G., S.M.M., T.H., C.-C.L., B.A.D.B., G.A.F.).,Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia (K.J.B.)
| | - Owen Tang
- From the University of Sydney, Kolling Institute of Medical Research, Cardiothoracic and Vascular Health (K.J.B., O.T., C.G., S.M.M., T.H., C.-C.L., B.A.D.B., G.A.F.).,Royal North Shore Hospital, St Leonards, NSW, Australia (O.T., T.H., C.-C.L., B.A.D.B., G.A.F.)
| | - Carmine Gentile
- From the University of Sydney, Kolling Institute of Medical Research, Cardiothoracic and Vascular Health (K.J.B., O.T., C.G., S.M.M., T.H., C.-C.L., B.A.D.B., G.A.F.).,University of Technology Sydney, Ultimo, NSW, Australia (C.G., S.M.M.)
| | - Seyed M Moosavi
- From the University of Sydney, Kolling Institute of Medical Research, Cardiothoracic and Vascular Health (K.J.B., O.T., C.G., S.M.M., T.H., C.-C.L., B.A.D.B., G.A.F.).,University of Technology Sydney, Ultimo, NSW, Australia (C.G., S.M.M.)
| | - Thomas Hansen
- From the University of Sydney, Kolling Institute of Medical Research, Cardiothoracic and Vascular Health (K.J.B., O.T., C.G., S.M.M., T.H., C.-C.L., B.A.D.B., G.A.F.).,Royal North Shore Hospital, St Leonards, NSW, Australia (O.T., T.H., C.-C.L., B.A.D.B., G.A.F.)
| | - Chia-Chi Liu
- From the University of Sydney, Kolling Institute of Medical Research, Cardiothoracic and Vascular Health (K.J.B., O.T., C.G., S.M.M., T.H., C.-C.L., B.A.D.B., G.A.F.).,Royal North Shore Hospital, St Leonards, NSW, Australia (O.T., T.H., C.-C.L., B.A.D.B., G.A.F.).,Heart Research Institute, Newtown, NSW, Australia (C.-C.L.)
| | - Belinda A Di Bartolo
- From the University of Sydney, Kolling Institute of Medical Research, Cardiothoracic and Vascular Health (K.J.B., O.T., C.G., S.M.M., T.H., C.-C.L., B.A.D.B., G.A.F.).,Royal North Shore Hospital, St Leonards, NSW, Australia (O.T., T.H., C.-C.L., B.A.D.B., G.A.F.)
| | - Gemma A Figtree
- From the University of Sydney, Kolling Institute of Medical Research, Cardiothoracic and Vascular Health (K.J.B., O.T., C.G., S.M.M., T.H., C.-C.L., B.A.D.B., G.A.F.).,Royal North Shore Hospital, St Leonards, NSW, Australia (O.T., T.H., C.-C.L., B.A.D.B., G.A.F.)
| |
Collapse
|
35
|
Nephrotoxicity of Anti-Angiogenic Therapies. Diagnostics (Basel) 2021; 11:diagnostics11040640. [PMID: 33916159 PMCID: PMC8066213 DOI: 10.3390/diagnostics11040640] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/19/2021] [Accepted: 03/29/2021] [Indexed: 12/24/2022] Open
Abstract
The use of inhibitors of vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor 2 (VEGFR2) signaling for the treatment of cancer has increased over the last decade. This signaling pathway plays a fundamental role in angiogenesis and also in kidney physiology. The emergence of anti-angiogenic therapies has led to adverse nephrotoxic effects, despite improving the outcomes of patients. In this review, we will present the different anti-angiogenic therapies targeting the VEGFR pathway in association with the incidence of renal manifestations during their use. In addition, we will discuss, in detail, the pathophysiological mechanisms of frequent renal diseases such as hypertension, proteinuria, renal dysfunction, and electrolyte disorders. Finally, we will outline the cellular damage described following these therapies.
Collapse
|
36
|
Maissan P, Mooij EJ, Barberis M. Sirtuins-Mediated System-Level Regulation of Mammalian Tissues at the Interface between Metabolism and Cell Cycle: A Systematic Review. BIOLOGY 2021; 10:194. [PMID: 33806509 PMCID: PMC7999230 DOI: 10.3390/biology10030194] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 02/06/2023]
Abstract
Sirtuins are a family of highly conserved NAD+-dependent proteins and this dependency links Sirtuins directly to metabolism. Sirtuins' activity has been shown to extend the lifespan of several organisms and mainly through the post-translational modification of their many target proteins, with deacetylation being the most common modification. The seven mammalian Sirtuins, SIRT1 through SIRT7, have been implicated in regulating physiological responses to metabolism and stress by acting as nutrient sensors, linking environmental and nutrient signals to mammalian metabolic homeostasis. Furthermore, mammalian Sirtuins have been implicated in playing major roles in mammalian pathophysiological conditions such as inflammation, obesity and cancer. Mammalian Sirtuins are expressed heterogeneously among different organs and tissues, and the same holds true for their substrates. Thus, the function of mammalian Sirtuins together with their substrates is expected to vary among tissues. Any therapy depending on Sirtuins could therefore have different local as well as systemic effects. Here, an introduction to processes relevant for the actions of Sirtuins, such as metabolism and cell cycle, will be followed by reasoning on the system-level function of Sirtuins and their substrates in different mammalian tissues. Their involvement in the healthy metabolism and metabolic disorders will be reviewed and critically discussed.
Collapse
Affiliation(s)
- Parcival Maissan
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
| | - Eva J. Mooij
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| | - Matteo Barberis
- Synthetic Systems Biology and Nuclear Organization, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 XH Amsterdam, The Netherlands;
- Systems Biology, School of Biosciences and Medicine, Faculty of Health and Medical Sciences, University of Surrey, Guildford GU2 7XH, Surrey, UK;
- Centre for Mathematical and Computational Biology, CMCB, University of Surrey, Guildford GU2 7XH, Surrey, UK
| |
Collapse
|
37
|
Pugazhendhi A, Hubbell M, Jairam P, Ambati B. Neovascular Macular Degeneration: A Review of Etiology, Risk Factors, and Recent Advances in Research and Therapy. Int J Mol Sci 2021; 22:1170. [PMID: 33504013 PMCID: PMC7866170 DOI: 10.3390/ijms22031170] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/11/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
Neovascular age-related macular degeneration (exudative or wet AMD) is a prevalent, progressive retinal degenerative macular disease that is characterized by neovascularization of the choroid, mainly affecting the elderly population causing gradual vision impairment. Risk factors such as age, race, genetics, iris color, smoking, drinking, BMI, and diet all play a part in nvAMD's progression, with anti-vascular endothelial growth factor (anti-VEGF) therapy being the mainstay of treatment. Current therapeutic advancements slow the progression of the disease but do not cure or reverse its course. Newer therapies such as gene therapies, Rho-kinase inhibitors, and levodopa offer potential new targets for treatment.
Collapse
Affiliation(s)
- Arunbalaji Pugazhendhi
- Knights Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR 97403, USA; (A.P.); (M.H.)
| | - Margaret Hubbell
- Knights Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR 97403, USA; (A.P.); (M.H.)
| | - Pooja Jairam
- Vagelos College of Physicians & Surgeons, Columbia Irving Medical Center, Columbia University, New York, NY 10032, USA;
| | - Balamurali Ambati
- Knights Campus for Accelerating Scientific Impact, University of Oregon, Eugene, OR 97403, USA; (A.P.); (M.H.)
| |
Collapse
|
38
|
Gil CL, Hooker E, Larrivée B. Diabetic Kidney Disease, Endothelial Damage, and Podocyte-Endothelial Crosstalk. Kidney Med 2021; 3:105-115. [PMID: 33604542 PMCID: PMC7873832 DOI: 10.1016/j.xkme.2020.10.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Diabetes-related complications are a significant source of morbidity and mortality worldwide. Diabetic kidney disease is a frequent microvascular complication and a primary cause of kidney failure in patients with diabetes. The glomerular filtration barrier is composed of 3 layers: the endothelium, glomerular basement membrane, and podocytes. Podocytes and the endothelium communicate through molecular crosstalk to maintain filtration at the glomerular filtration barrier. Chronic hyperglycemia affects all 3 layers of the glomerular filtration barrier, as well as the molecular crosstalk that occurs between the 2 cellular layers. One of the earliest events following chronic hyperglycemia is endothelial cell dysfunction. Early endothelial damage is associated with progression of diabetic kidney disease. However, current therapies are based in controlling glycemia and arterial blood pressure without targeting endothelial dysfunction. Disruption of the endothelial cell layer also alters the molecular crosstalk that occurs between the endothelium and podocytes. This review discusses both the physiologic and pathologic communication that occurs at the glomerular filtration barrier. It examines how these signaling components contribute to podocyte foot effacement, podocyte detachment, and the progression of diabetic kidney disease.
Collapse
Affiliation(s)
- Cindy Lora Gil
- Department of Biomedical Sciences, University of Montreal, Montréal, QC, Canada
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montréal, QC, Canada
| | - Erika Hooker
- Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montréal, QC, Canada
| | - Bruno Larrivée
- Department of Ophtalmology, University of Montreal, Montréal, QC, Canada
| |
Collapse
|
39
|
Lassén E, Daehn IS. Molecular Mechanisms in Early Diabetic Kidney Disease: Glomerular Endothelial Cell Dysfunction. Int J Mol Sci 2020; 21:ijms21249456. [PMID: 33322614 PMCID: PMC7764016 DOI: 10.3390/ijms21249456] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/06/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease (ESRD), with prevalence increasing at an alarming rate worldwide and today, there are no known cures. The pathogenesis of DKD is complex, influenced by genetics and the environment. However, the underlying molecular mechanisms that contribute to DKD risk in about one-third of diabetics are still poorly understood. The early stage of DKD is characterized by glomerular hyperfiltration, hypertrophy, podocyte injury and depletion. Recent evidence of glomerular endothelial cell injury at the early stage of DKD has been suggested to be critical in the pathological process and has highlighted the importance of glomerular intercellular crosstalk. A potential mechanism may include reactive oxygen species (ROS), which play a direct role in diabetes and its complications. In this review, we discuss different cellular sources of ROS in diabetes and a new emerging paradigm of endothelial cell dysfunction as a key event in the pathogenesis of DKD.
Collapse
|
40
|
Reina-Torres E, De Ieso ML, Pasquale LR, Madekurozwa M, van Batenburg-Sherwood J, Overby DR, Stamer WD. The vital role for nitric oxide in intraocular pressure homeostasis. Prog Retin Eye Res 2020; 83:100922. [PMID: 33253900 DOI: 10.1016/j.preteyeres.2020.100922] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 11/13/2020] [Accepted: 11/23/2020] [Indexed: 02/07/2023]
Abstract
Catalyzed by endothelial nitric oxide (NO) synthase (eNOS) activity, NO is a gaseous signaling molecule maintaining endothelial and cardiovascular homeostasis. Principally, NO regulates the contractility of vascular smooth muscle cells and permeability of endothelial cells in response to either biochemical or biomechanical cues. In the conventional outflow pathway of the eye, the smooth muscle-like trabecular meshwork (TM) cells and Schlemm's canal (SC) endothelium control aqueous humor outflow resistance, and therefore intraocular pressure (IOP). The mechanisms by which outflow resistance is regulated are complicated, but NO appears to be a key player as enhancement or inhibition of NO signaling dramatically affects outflow function; and polymorphisms in NOS3, the gene that encodes eNOS modifies the relation between various environmental exposures and glaucoma. Based upon a comprehensive review of past foundational studies, we present a model whereby NO controls a feedback signaling loop in the conventional outflow pathway that is sensitive to changes in IOP and its oscillations. Thus, upon IOP elevation, the outflow pathway tissues distend, and the SC lumen narrows resulting in increased SC endothelial shear stress and stretch. In response, SC cells upregulate the production of NO, relaxing neighboring TM cells and increasing permeability of SC's inner wall. These IOP-dependent changes in the outflow pathway tissues reduce the resistance to aqueous humor drainage and lower IOP, which, in turn, diminishes the biomechanical signaling on SC. Similar to cardiovascular pathogenesis, dysregulation of the eNOS/NO system leads to dysfunctional outflow regulation and ocular hypertension, eventually resulting in primary open-angle glaucoma.
Collapse
Affiliation(s)
| | | | - Louis R Pasquale
- Eye and Vision Research Institute of New York Eye and Ear Infirmary at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Darryl R Overby
- Department of Bioengineering, Imperial College London, London, UK.
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University, Durham, NC, USA.
| |
Collapse
|
41
|
Sol M, Kamps JAAM, van den Born J, van den Heuvel MC, van der Vlag J, Krenning G, Hillebrands JL. Glomerular Endothelial Cells as Instigators of Glomerular Sclerotic Diseases. Front Pharmacol 2020; 11:573557. [PMID: 33123011 PMCID: PMC7573930 DOI: 10.3389/fphar.2020.573557] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/14/2020] [Indexed: 12/20/2022] Open
Abstract
Glomerular endothelial cell (GEnC) dysfunction is important in the pathogenesis of glomerular sclerotic diseases, including Focal Segmental Glomerulosclerosis (FSGS) and overt diabetic nephropathy (DN). GEnCs form the first cellular barrier in direct contact with cells and factors circulating in the blood. Disturbances in these circulating factors can induce GEnC dysfunction. GEnC dysfunction occurs in early stages of FSGS and DN, and is characterized by a compromised endothelial glycocalyx, an inflammatory phenotype, mitochondrial damage and oxidative stress, aberrant cell signaling, and endothelial-to-mesenchymal transition (EndMT). GEnCs are in an interdependent relationship with podocytes and mesangial cells, which involves bidirectional cross-talk via intercellular signaling. Given that GEnC behavior directly influences podocyte function, it is conceivable that GEnC dysfunction may culminate in podocyte damage, proteinuria, subsequent mesangial activation, and ultimately glomerulosclerosis. Indeed, GEnC dysfunction is sufficient to cause podocyte injury, proteinuria and activation of mesangial cells. Aberrant gene expression patterns largely contribute to GEnC dysfunction and epigenetic changes seem to be involved in causing aberrant transcription. This review summarizes literature that uncovers the importance of cross-talk between GEnCs and podocytes, and GEnCs and mesangial cells in the context of the development of FSGS and DN, and the potential use of GEnCs as efficacious cellular target to pharmacologically halt development and progression of DN and FSGS.
Collapse
Affiliation(s)
- Marloes Sol
- Department of Pathology and Medical Biology, Division of Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jan A A M Kamps
- Department of Pathology and Medical Biology, Division of Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jacob van den Born
- Department of Internal Medicine, Division of Nephrology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Marius C van den Heuvel
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Johan van der Vlag
- Department of Nephrology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Guido Krenning
- Department of Pathology and Medical Biology, Division of Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| |
Collapse
|
42
|
Pal S, Rashid M, Singh SK, Porwal K, Singh P, Mohamed R, Gayen JR, Wahajuddin M, Chattopadhyay N. Skeletal restoration by phosphodiesterase 5 inhibitors in osteopenic mice: Evidence of osteoanabolic and osteoangiogenic effects of the drugs. Bone 2020; 135:115305. [PMID: 32126313 DOI: 10.1016/j.bone.2020.115305] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 02/12/2020] [Accepted: 02/28/2020] [Indexed: 12/16/2022]
Abstract
Phosphodiesterases (PDEs) hydrolyze cyclic nucleotides and thereby regulate diverse cellular functions. The reports on the skeletal effects of PDE inhibitors are conflicting. Here, we screened 17 clinically used non-xanthine PDE inhibitors (selective and non-selective) using mouse calvarial osteoblasts (MCO) where the readout was osteoblast differentiation. From this screen, we identified sildenafil and vardenafil (both PDE5 inhibitors) having the least osteogenic EC50. Both drugs significantly increased vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2) expressions in MCO and the nitric oxide synthase inhibitor L-NAME completely blocked VEGF expression induced by these drugs. Sunitinib, a tyrosine receptor kinase inhibitor that also blocks VEGFR2 blocked sildenafil-/vardenafil-induced osteoblast differentiation. At half of their human equivalent doses, i.e. 6.0 mg/kg sildenafil and 2.5 mg/kg vardenafil, the maximum bone marrow level of sildenafil was 32% and vardenafil was 21% of their blood levels. At these doses, both drugs enhanced bone regeneration at the femur osteotomy site and completely restored bone mass, microarchitecture, and strength in OVX mice. Furthermore, both drugs increased surface referent bone formation and serum bone formation marker (P1NP) without affecting the resorption marker (CTX-1). Both drugs increased the expression of VEGF and VEGFR2 in bones and osteoblasts and increased skeletal vascularity. Sunitinib completely blocked the bone restorative and vascular effects of sildenafil and vardenafil in OVX mice. Taken together, our study suggested that sildenafil and vardenafil at half of their adult human doses completely reversed osteopenia in OVX mice by an osteogenic mechanism that was associated with enhanced skeletal vascularity.
Collapse
Affiliation(s)
- Subhashis Pal
- Division of Endocrinology, Central Drug Research Institute (CDRI), Council of Scientific and Industrial Research (CSIR), Lucknow 226031, India
| | - Mamunur Rashid
- Pharmaceutics & Pharmacokinetics Division, CDRI-CSIR, Lucknow 226031, India
| | | | - Konica Porwal
- Division of Endocrinology, Central Drug Research Institute (CDRI), Council of Scientific and Industrial Research (CSIR), Lucknow 226031, India
| | - Priya Singh
- Division of Endocrinology, Central Drug Research Institute (CDRI), Council of Scientific and Industrial Research (CSIR), Lucknow 226031, India
| | - Riyazuddin Mohamed
- Pharmaceutics & Pharmacokinetics Division, CDRI-CSIR, Lucknow 226031, India
| | - Jiaur R Gayen
- Pharmaceutics & Pharmacokinetics Division, CDRI-CSIR, Lucknow 226031, India
| | | | - Naibedya Chattopadhyay
- Division of Endocrinology, Central Drug Research Institute (CDRI), Council of Scientific and Industrial Research (CSIR), Lucknow 226031, India.
| |
Collapse
|
43
|
Salie R, Alsalhin AKH, Marais E, Lochner A. Cardioprotective Effects of Beta3-Adrenergic Receptor (β3-AR) Pre-, Per-, and Post-treatment in Ischemia-Reperfusion. Cardiovasc Drugs Ther 2020; 33:163-177. [PMID: 30729348 DOI: 10.1007/s10557-019-06861-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The β3-AR (beta3-adrenergic receptor) is resistant to short-term agonist-promoted desensitization and delivers a constant intracellular signal, making this receptor a potential target in acute myocardial infarction (AMI). AIM To investigate whether selective modulation of β3-AR prior to or during ischemia and/or reperfusion may be cardioprotective. METHODS Isolated perfused rat hearts were exposed to 35-min regional ischemia (RI) and 60-min reperfusion. The β3-AR agonist (BRL37344, 1 μM) or antagonist (SR59230A, 0.1 μM) was applied: (i) before RI (PreT) or (ii) last 10 min of RI (PerT) or (iii) onset of reperfusion (PostT) or (iv) during both PerT+PostT. Nitric oxide (NO) involvement was assessed, using the NOS inhibitor, L-NAME (50 μM). Endpoints were functional recovery, infarct size (IS), cGMP levels, and Western blot analysis of eNOS, ERKp44/p42, PKB/Akt, and glycogen synthase kinase-3β (GSK-3β). RESULTS Selective treatment with BRL significantly reduced IS. L-NAME abolished BRL-mediated cardioprotection. BRL (PreT) and BRL (PerT) significantly increased cGMP levels (which were reduced by L-NAME) and PKB/Akt phosphorylation. BRL (PostT) produced significantly increased cGMP levels, PKB/Akt, and ERKp44/p42 phosphorylation. BRL (PerT+PostT) caused significant eNOS, PKB/Akt, ERKp44/p42, and GSK-3β phosphorylation. CONCLUSION β3-AR activation by BRL37344 induced significant cardioprotection regardless of the experimental protocol. However, the pattern of intracellular signaling with each BRL treatment differed to some degree and suggests the involvement of cGMP, eNOS, ERK, GSK-3β, and particularly PKB/Akt activation. The data also suggest that clinical application of β3-AR stimulation should preferably be incorporated during late ischemia or/and early reperfusion.
Collapse
Affiliation(s)
- Ruduwaan Salie
- Biomedical Research and Innovation Platform, South African Medical Research Council, Building D, Medicina, Francie van Zijl Drive, Parow Valley, Cape Town, Western Cape, South Africa.
- Faculty of Medicine and Health Sciences, Division of Medical Physiology, University of Stellenbosch, PO Box 19063, Cape Town, South Africa.
| | - Aisha Khlani Hassan Alsalhin
- Faculty of Medicine and Health Sciences, Division of Medical Physiology, University of Stellenbosch, PO Box 19063, Cape Town, South Africa
| | - Erna Marais
- Faculty of Medicine and Health Sciences, Division of Medical Physiology, University of Stellenbosch, PO Box 19063, Cape Town, South Africa
| | - Amanda Lochner
- Faculty of Medicine and Health Sciences, Division of Medical Physiology, University of Stellenbosch, PO Box 19063, Cape Town, South Africa
| |
Collapse
|
44
|
Deluque AL, de Almeida LF, Francescato HDC, da Silva CGA, Costa RS, Antunes-Rodrigues J, Coimbra TM. Effect of Calcitriol on the Renal Microvasculature Differentiation Disturbances Induced by AT 1 Blockade During Nephrogenesis in Rats. Front Med (Lausanne) 2020; 7:23. [PMID: 32118008 PMCID: PMC7016013 DOI: 10.3389/fmed.2020.00023] [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: 10/27/2019] [Accepted: 01/15/2020] [Indexed: 12/11/2022] Open
Abstract
Alterations in the renal vasculature during fetal programming can cause disturbances in renal structure and function that persist into adulthood. Calcitriol can affect cellular differentiation and proliferation, and promote endothelial cell maintenance, each of which is a key event in nephrogenesis. Calcitriol is a negative endocrine regulator of the renin gene. Rats exposed to renin-angiotensin system (RAS) antagonists during lactation have been shown to develop renal disorders, which demonstrated that the RAS may play an important role in mammalian kidney development. We evaluated the effects of calcitriol administration on losartan [angiotensin II receptor antagonist (ANGII), AT1]-induced changes in renal differentiation in rats during lactation. Rats treated with losartan showed alterations in renal function and structure that persisted into adulthood. These disruptions included hydronephrosis, papillary atrophy, endothelial dysfunction, and aberrant endothelial structure. These changes were mitigated by treatment with calcitriol. The results of our study showed that animals exposed to AT1 blockade during lactation exhibited altered renal microvasculature differentiation in adulthood that was attenuated by treatment with calcitriol.
Collapse
Affiliation(s)
- Amanda L Deluque
- Laboratory of Renal Physiology, Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Lucas F de Almeida
- Laboratory of Renal Physiology, Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Heloísa D C Francescato
- Laboratory of Renal Physiology, Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Cleonice G A da Silva
- Laboratory of Renal Physiology, Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Roberto S Costa
- Laboratory of Renal Pathology, Division of Nephrology, Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - José Antunes-Rodrigues
- Laboratory of Neuroendocrinology, Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| | - Terezila M Coimbra
- Laboratory of Renal Physiology, Department of Physiology, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
45
|
Addis DR, Lambert JA, Ren C, Doran S, Aggarwal S, Jilling T, Matalon S. Vascular Endothelial Growth Factor-121 Administration Mitigates Halogen Inhalation-Induced Pulmonary Injury and Fetal Growth Restriction in Pregnant Mice. J Am Heart Assoc 2020; 9:e013238. [PMID: 32009528 PMCID: PMC7033856 DOI: 10.1161/jaha.119.013238] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 01/07/2020] [Indexed: 12/16/2022]
Abstract
Background Circulating levels of sFLT-1 (soluble fms-like tyrosine kinase 1), the extracellular domain of vascular endothelial growth factor (VEGF) receptor 1, and its ratio to levels of placental growth factor are markers of the occurrence and severity of preeclampsia. Methods and Results C57BL/6 pregnant mice on embryonic day 14.5 (E14.5), male, and non-pregnant female mice were exposed to air or to Br2 at 600 ppm for 30 minutes and were treated with vehicle or with VEGF-121 (100 μg/kg, subcutaneously) daily, starting 48 hours post-exposure. Plasma, bronchoalveolar lavage fluid, lungs, fetuses, and placentas were collected 120 hours post-exposure. In Br2-exposed pregnant mice, there was a time-dependent and significant increase in plasma levels of sFLT-1 which correlated with increases in mouse lung wet/dry weights and bronchoalveolar lavage fluid protein content. Supplementation of exogenous VEGF-121 improved survival and weight gain, reduced lung wet/dry weights, decreased bronchoalveolar lavage fluid protein levels, enhanced placental development, and improved fetal growth in pregnant mice exposed to Br2. Exogenous VEGF-121 administration had no effect in non-pregnant mice. Conclusions These results implicate inhibition of VEGF signaling driven by sFLT-1 overexpression as a mechanism of pregnancy-specific injury leading to lung edema, maternal mortality, and fetal growth restriction after bromine gas exposure.
Collapse
Affiliation(s)
- Dylan R. Addis
- Division of Cardiothoracic AnesthesiologyDepartment of Anesthesiology and Perioperative MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- Division of Molecular and Translational BiomedicineDepartment of Anesthesiology and Perioperative MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- UAB Comprehensive Cardiovascular CenterUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| | - James A. Lambert
- Division of Molecular and Translational BiomedicineDepartment of Anesthesiology and Perioperative MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- Graduate Biomedical SciencesBiochemistry, Structural and Stem Cell Biology ThemeUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| | - Changchun Ren
- Department of PediatricsDivision of NeonatologyUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| | - Stephen Doran
- Division of Molecular and Translational BiomedicineDepartment of Anesthesiology and Perioperative MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| | - Saurabh Aggarwal
- Division of Molecular and Translational BiomedicineDepartment of Anesthesiology and Perioperative MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| | - Tamas Jilling
- Department of PediatricsDivision of NeonatologyUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| | - Sadis Matalon
- Division of Molecular and Translational BiomedicineDepartment of Anesthesiology and Perioperative MedicineUniversity of Alabama at BirminghamBirminghamALUSA
- UAB Comprehensive Cardiovascular CenterUniversity of Alabama at BirminghamBirminghamALUSA
- University of Alabama School of MedicineBirminghamALUSA
| |
Collapse
|
46
|
Liu Y, Paterson M, Baumgardt SL, Irwin MG, Xia Z, Bosnjak ZJ, Ge ZD. Vascular endothelial growth factor regulation of endothelial nitric oxide synthase phosphorylation is involved in isoflurane cardiac preconditioning. Cardiovasc Res 2020; 115:168-178. [PMID: 29931049 DOI: 10.1093/cvr/cvy157] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 06/19/2018] [Indexed: 12/13/2022] Open
Abstract
Aims Previous studies indicate that nitric oxide derived from endothelial nitric oxide synthase (eNOS) serves as both trigger and mediator in anaesthetic cardiac preconditioning. The mechanisms underlying regulation of eNOS by volatile anaesthetics have not been fully understood. Therefore, this study examined the role of vascular endothelial growth factor (VEGF) in isoflurane cardiac preconditioning. Methods and results Wistar rats underwent 30 min of coronary artery occlusion followed by 2 h of reperfusion. Isoflurane given prior to ischaemia/reperfusion significantly decreased myocardial infarct size from 60 ± 1% in control to 40 ± 3% (n = 8 rats/group, P < 0.05). The beneficial effects of isoflurane were blocked by neutralizing antibody against VEGF (nVEGF). Coronary arterial endothelial cells (ECs) alone or together with cardiomyocytes (CMs) were subjected to hypoxia/reoxygenation injury. The expression of VEGF and eNOS was analysed by western blot, and nitric oxide was measured by ozone-based chemiluminescence. In co-cultured CMs and ECs, isoflurane administered before hypoxia/reoxygenation attenuated lactate dehydrogenase activity and increased the ratio of phosphorylated eNOS/eNOS and nitric oxide production. The protective effect of isoflurane on CMs was compromised by nVEGF and after VEGF in ECs was inhibited with hypoxia inducible factor-1α short hairpin RNA (shRNA). The negative effect of hypoxia inducible factor-1α shRNA was restored by recombinant VEGF. Conclusion Isoflurane cardiac preconditioning is associated with VEGF regulation of phosphorylation of eNOS and nitric oxide production.
Collapse
Affiliation(s)
- Yanan Liu
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA.,Department of Pathology and Cell Biology, Columbia University, 630 W. 168th Street, New York, NY, USA.,Department of Anesthesiology, University of Hong Kong, Hong Kong, China SAR, China
| | - Mark Paterson
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA
| | - Shelley L Baumgardt
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA
| | - Michael G Irwin
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China SAR, China
| | - Zhengyuan Xia
- Department of Anesthesiology, University of Hong Kong, Hong Kong, China SAR, China
| | - Zeljko J Bosnjak
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA.,Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA
| | - Zhi-Dong Ge
- Department of Anesthesiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, USA.,Department of Ophthalmology, Stanford University School of Medicine, 1651 Page Mill Road, Stanford, CA, USA
| |
Collapse
|
47
|
Hulin JA, Gubareva EA, Jarzebska N, Rodionov RN, Mangoni AA, Tommasi S. Inhibition of Dimethylarginine Dimethylaminohydrolase (DDAH) Enzymes as an Emerging Therapeutic Strategy to Target Angiogenesis and Vasculogenic Mimicry in Cancer. Front Oncol 2020; 9:1455. [PMID: 31993367 PMCID: PMC6962312 DOI: 10.3389/fonc.2019.01455] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/05/2019] [Indexed: 01/01/2023] Open
Abstract
The small free radical gas nitric oxide (NO) plays a key role in various physiological and pathological processes through enhancement of endothelial cell survival and proliferation. In particular, NO has emerged as a molecule of interest in carcinogenesis and tumor progression due to its crucial role in various cancer-related events including cell invasion, metastasis, and angiogenesis. The dimethylarginine dimethylaminohydrolase (DDAH) family of enzymes metabolize the endogenous nitric oxide synthase (NOS) inhibitors, asymmetric dimethylarginine (ADMA) and monomethyl arginine (L-NMMA), and are thus key for maintaining homeostatic control of NO. Dysregulation of the DDAH/ADMA/NO pathway resulting in increased local NO availability often promotes tumor growth, angiogenesis, and vasculogenic mimicry. Recent literature has demonstrated increased DDAH expression in tumors of different origins and has also suggested a potential ADMA-independent role for DDAH enzymes in addition to their well-studied ADMA-mediated influence on NO. Inhibition of DDAH expression and/or activity in cell culture models and in vivo studies has indicated the potential therapeutic benefit of this pathway through inhibition of both angiogenesis and vasculogenic mimicry, and strategies for manipulating DDAH function in cancer are currently being actively pursued by several research groups. This review will thus provide a timely discussion on the expression, regulation, and function of DDAH enzymes in regard to angiogenesis and vasculogenic mimicry, and will offer insight into the therapeutic potential of DDAH inhibition in cancer based on preclinical studies.
Collapse
Affiliation(s)
- Julie-Ann Hulin
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Ekaterina A Gubareva
- N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg, Russia
| | - Natalia Jarzebska
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Anesthesiology and Intensive Care Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Roman N Rodionov
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Arduino A Mangoni
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Sara Tommasi
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
48
|
Nasirzadeh M, Rasmi Y, Rahbarghazi R, Kheradmand F, Karimipour M, Aramwit P, Astinfeshan M, Gholinejad Z, Daeihasani B, Saboory E, Shirpoor A, Rezabakhsh A, Zolali E, Khalaji N. Crocetin promotes angiogenesis in human endothelial cells through PI3K-Akt-eNOS signaling pathway. EXCLI JOURNAL 2019; 18:936-949. [PMID: 31762720 PMCID: PMC6868919 DOI: 10.17179/excli2019-1175] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 10/14/2019] [Indexed: 02/02/2023]
Abstract
Previous studies proved the pro-angiogenic effect of Crocetin, a natural carotenoid dicarboxylic acid, in both in vivo and in vitro models. However, the exact mechanism of Crocetin action has not completely been elucidated yet. The current experiment was designed to find the activity of PI3K-Akt-eNOS axis after the treatment of endothelial cells with Crocetin in vitro. Human Umbilical Vein Endothelial Cells (HUVECs) were incubated with various concentrations of Crocetin (1, 5, 25, 50, and 100 µM) over a period of 72 h. Crocetin significantly increased HUVECs viability after 72 h as compared with the control group. We also found that Crocetin promoted the formation of the capillary-like structure compared to the control (p<0.05). Moreover, an improved migration rate and increased MMP-9 activity were observed in HUVECs that received 50 µM Crocetin (p<0.05). Crocetin enhanced the uptake of Ac-LDL which is correlated with increased lipid metabolism. Based on the data from the current experiment, protein level of VEGFR-1, -2 and p-Akt/Akt, p-eNOS/eNOS ratios were increased 72 h after the treatment of HUVECs with Crocetin (p<0.05). In contrast, the transcription level of VEGF was reduced in Crocetin-treated cells. These data demonstrated that Crocetin promotes HUVECs angiogenesis potential by the modulation of VEGF signaling pathway and increased cell viability. The PI3K/Akt/eNOS axis is required for a Crocetin-associated activity in endothelial cells.
Collapse
Affiliation(s)
- Mahdieh Nasirzadeh
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yousef Rasmi
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.,Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Reza Rahbarghazi
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Kheradmand
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mojtaba Karimipour
- Department of Anatomy, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Pornanong Aramwit
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Phaya Thai Road, Phatumwan, Bangkok 10330, Thailand
| | - Maryam Astinfeshan
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Zafar Gholinejad
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Behrokh Daeihasani
- Department of Biology, Payame Noor University, P.O.Box 19395-3697, Tehran, Iran
| | - Ehsan Saboory
- Neuroscience Research Center, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Alireza Shirpoor
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Aysa Rezabakhsh
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elmira Zolali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Naser Khalaji
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
49
|
Wang Y, Lu YH, Tang C, Xue M, Li XY, Chang YP, Cheng Y, Li T, Yu XC, Sun B, Li CJ, Chen LM. Calcium Dobesilate Restores Autophagy by Inhibiting the VEGF/PI3K/AKT/mTOR Signaling Pathway. Front Pharmacol 2019; 10:886. [PMID: 31447680 PMCID: PMC6696883 DOI: 10.3389/fphar.2019.00886] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/15/2019] [Indexed: 01/10/2023] Open
Abstract
Objective: Calcium dobesilate (CaD), an effective drug for the treatment of diabetic microvascular complications, especially diabetic retinopathy, is widely used in the clinic. Interestingly, several studies have indicated that CaD is therapeutic for diabetic kidney disease (DKD). Recently, evidence has indicated that altered vascular endothelial growth factor (VEGF) expression and decreased autophagy are the main pathological mechanisms of proteinuria. Thus, this study was conducted to explore the effect of CaD on restoring autophagy in DKD and the possible signaling pathway between VEGF and autophagy. Methods: Obese mice with spontaneous diabetes (KK-Ay) and high-fat diet- and streptozotocin-induced diabetic mice (HFD/STZ) were used in this study. Biochemical staining, western blotting, and immunohistochemistry were conducted to determine the angioprotective effect of CaD and the underlying mechanism between autophagy and VEGF/VEGFR. Results: Our results showed that CaD was capable of reducing albuminuria and restoring renal histological changes in KK-Ay and HFD/STZ-induced diabetic mice. CaD restored autophagy by decreasing the protein expression of LC3 II, Atg5, and beclin 1 and increasing the expression of P62. Moreover, CaD reduced the activation of the autophagy-related PI3K/AKT/mTOR pathway possibly via decreasing VEGF and downregulating VEGF receptor 2. Conclusion: Overall, CaD, as a novel potential therapeutic drug for DKD, plays a key role in protecting renal function and restoring autophagy by blocking VEGF/VEGFR2 and inhibiting the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yue Wang
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, China
| | - Yun-Hong Lu
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, China
| | - Chao Tang
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, China
| | - Mei Xue
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, China
| | - Xiao-Yu Li
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, China
| | - Yun-Peng Chang
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, China
| | - Ying Cheng
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, China
| | - Ting Li
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, China
| | - Xiao-Chen Yu
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, China
| | - Bei Sun
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, China
| | - Chun-Jun Li
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, China
| | - Li-Ming Chen
- NHC Key Laboratory of Hormones and Development (Tianjin Medical University), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University Chu Hsien-I Memorial Hospital, Tianjin Institute of Endocrinology, Tianjin, China
| |
Collapse
|
50
|
Plummer C, Michael A, Shaikh G, Stewart M, Buckley L, Miles T, Ograbek A, McCormack T. Expert recommendations on the management of hypertension in patients with ovarian and cervical cancer receiving bevacizumab in the UK. Br J Cancer 2019; 121:109-116. [PMID: 31182765 PMCID: PMC6738076 DOI: 10.1038/s41416-019-0481-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 03/15/2019] [Accepted: 05/02/2019] [Indexed: 01/09/2023] Open
Abstract
Bevacizumab is an anti-vascular endothelial growth factor monoclonal antibody that may prolong survival in ovarian and cervical cancer when given in combination with chemotherapy. It works by blocking the signalling pathways that are required for tumour angiogenesis, potentially limiting the cancer’s ability to grow and spread. Hypertension is a known side effect of all angiogenesis inhibitors and could lead to interruption or premature discontinuation of effective anti-cancer treatment. Hypertension may also act as a barrier to the initiation of such treatment. In this review, we aim to present clear and practical recommendations on the management of blood pressure in ovarian and cervical cancer patients before, during and after bevacizumab treatment. This guidance covers considerations before initiating bevacizumab therapy and recommendations on the management of patients who develop hypertension, or who experience worsening of pre-existing hypertension, during bevacizumab treatment, and once the course of bevacizumab has been completed. These recommendations were developed collaboratively by a group of clinicians, comprising cardiologists, oncologists, a general practitioner and specialist oncology nurses, with expertise and practical experience in either oncology or hypertension. The aim of these recommendations is to support oncologists with hypertension assessment and management to facilitate starting or continuing bevacizumab.
Collapse
Affiliation(s)
| | - Agnieszka Michael
- St Luke's Cancer Centre, Royal Surrey County Hospital, Guildford, UK
| | - Ghazia Shaikh
- Northern Centre for Cancer Care, Newcastle upon Tyne, UK
| | | | - Lynn Buckley
- Hull and East Yorkshire Hospitals NHS Trust, Cottingham, UK
| | - Tracie Miles
- Royal United Hospitals Bath NHS Foundation Trust, Bath, UK
| | | | | |
Collapse
|