1
|
Fu X, Yu B, Lu L, Han Y, Liu Y, Zhang J, Chen T, Yu D. An injectable and photocurable methacrylate-silk fibroin/prussian blue nanozyme hydrogel with antioxidant and pyroptosis suppression properties for cartilage regeneration. Int J Biol Macromol 2025; 307:142154. [PMID: 40118414 DOI: 10.1016/j.ijbiomac.2025.142154] [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: 10/21/2024] [Revised: 12/15/2024] [Accepted: 03/14/2025] [Indexed: 03/23/2025]
Abstract
Cartilage injury is one of the most prevalent, distressing, and disabling chronic conditions affecting degenerative joints worldwide; however, its underlying mechanisms remain elusive. Various biomaterials have been widely employed in the treatment of articular cartilage (AC) injuries. Despite these efforts, the key role of reactive oxygen species (ROS) as primary instigators of pyroptosis, combined with the lack of effective interventions, often results in suboptimal cartilage repair outcomes. Thus, there is an urgent need for the development of novel antioxidants and inflammasome-mediated pyroptosis modulators to create a favorable microenvironment for cartilage repair. In this study, we synthesized Prussian blue nanoparticles (PBNPs) capable of efficiently scavenging ROS. In vitro, these PBNPs protected against oxidative stress-induced cytotoxicity, preserved mitochondrial integrity, reduced the activation of nucleotide-binding domain and leucine-rich repeat family pyrin domain-containing 3 (NLRP3) inflammasomes and caspase-1, and subsequently downregulated gasdermin D (GSDMD) cleavage and inflammatory factor production, leading to the inhibition of chondrocyte pyroptosis. To extend these findings in vivo, we developed an injectable and photocurable methacrylate-silk fibroin (SilMA) hydrogel with homogeneously incorporated PBNPs, designed for releasing PBNPs. The resulting PBNPs@SilMA hydrogel decreased ROS production, reduced chondrocyte pyroptosis, and supported chondrocyte proliferation and matrix secretion, subsequently improved AC repair. Overall, our results indicate that the PBNPs@SilMA platform holds significant promise as a therapeutic strategy for AC injury.
Collapse
Affiliation(s)
- Xuefei Fu
- Department of Orthopedics, Anhui No.2 Provincial People's Hospital, Hefei 230041, China
| | - Bing Yu
- Department of Orthopedics, Anhui No.2 Provincial People's Hospital, Hefei 230041, China
| | - Lin Lu
- Department of Radiotherapy, Anhui No.2 Provincial People's Hospital, Hefei 230041, China
| | - Yafei Han
- Department of Orthopedics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Yancheng Liu
- Department of Bone Tumor and Soft Tissue Oncology, Tianjin Hospital, Tianjin University, Tianjin 300211, China
| | - Jingyu Zhang
- Department of Bone Tumor and Soft Tissue Oncology, Tianjin Hospital, Tianjin University, Tianjin 300211, China.
| | - Tao Chen
- Department of Orthopedics, Anhui No.2 Provincial People's Hospital, Hefei 230041, China.
| | - Defu Yu
- Department of Orthopedics, Anhui No.2 Provincial People's Hospital, Hefei 230041, China; School of Clinical Medicine, Anhui Medical College, China.
| |
Collapse
|
2
|
Bday J, Souid M, Pires V, Gabbouj S, Véjux A, Lizard G, Hassen E. Arginase Activity Inhibition With Thymoquinone Induces a Hybrid Type of Cell-Death in MDA-MB-231 Cell Line. J Biochem Mol Toxicol 2025; 39:e70130. [PMID: 39829401 PMCID: PMC11744436 DOI: 10.1002/jbt.70130] [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: 08/20/2024] [Revised: 11/18/2024] [Accepted: 12/29/2024] [Indexed: 01/22/2025]
Abstract
Arginase plays a crucial role in the urea cycle; it also has immunosuppressive and pro-tumor effects. The present study aimed to assess the effects of arginase inhibition by thymoquinone (2-Isopropyl-5-methyl-1,4-benzoquinone), an active compound of Nigella sativa, on cell death in the MDA-MB-231 triple-negative breast tumor cell line. Cell viability assays, Western blot analysis, and flow cytometry analysis were used to characterize oxidative stress and cell death. Our results showed that inhibition of arginase activity with thymoquinone significantly increased intracellular nitric oxide levels and resulted in overproduction of cellular and mitochondrial reactive oxygen species. Reductions in cell viability, cycle arrest, and increased cell death were also observed. Loss of transmembrane mitochondrial potential, activation of caspase-3, -7, and -9, cleavage of PARP, condensation and/or fragmentation of the nuclei, suggest that this cell death involved apoptosis. Furthermore, a cytoplasm vacuole formation and an increase in the ratio of [LC3-II/LC3-I] suggests a concomitant activation of autophagy with apoptosis. Altogether, the present study highlighted that arginase inhibition with thymoquinone induces a hybrid type of cell death defined as oxiapoptophagy. Thus, arginase inhibition with thymoquinone in the MDA-MB-231 cell line could be, in part, involved in the anticancer effect of thymoquinone.
Collapse
Affiliation(s)
- Jaweher Bday
- Molecular Immuno‐Oncology LaboratoryUniversity of MonastirMonastirTunisia
- Higher Institute of Biotechnology of MonastirUniversity of MonastirMonastirTunisia
| | - Moufida Souid
- Molecular Immuno‐Oncology LaboratoryUniversity of MonastirMonastirTunisia
- Higher Institute of Biotechnology of MonastirUniversity of MonastirMonastirTunisia
| | - Vivien Pires
- Bio‐PeroxIL ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ Laboratory (EA 7270)Université de Bourgogne (UB)DijonFrance
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut AgroUniversité de BourgogneDijonFrance
| | - Sallouha Gabbouj
- Molecular Immuno‐Oncology LaboratoryUniversity of MonastirMonastirTunisia
| | - Anne Véjux
- Bio‐PeroxIL ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ Laboratory (EA 7270)Université de Bourgogne (UB)DijonFrance
- Centre des Sciences du Goût et de l'Alimentation, CNRS, INRAE, Institut AgroUniversité de BourgogneDijonFrance
| | - Gérard Lizard
- Bio‐PeroxIL ‘Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism’ Laboratory (EA 7270)Université de Bourgogne (UB)DijonFrance
| | - Elham Hassen
- Molecular Immuno‐Oncology LaboratoryUniversity of MonastirMonastirTunisia
- Higher Institute of Biotechnology of MonastirUniversity of MonastirMonastirTunisia
| |
Collapse
|
3
|
Souid M, Bday J, Souissi S, Ghedira R, Gabbouj S, Shini-Hadhri S, Toumi D, Bergaoui H, Zouari I, Faleh R, Zakhama A, Hassen E. Arginase is upregulated in healthy women infected by oncogenic HPV types. Biomarkers 2023; 28:628-636. [PMID: 37860844 DOI: 10.1080/1354750x.2023.2273226] [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: 05/30/2023] [Accepted: 10/15/2023] [Indexed: 10/21/2023]
Abstract
INTRODUCTION The implication of arginase enzyme in Human Papillomavirus (HPV) infections has not been clearly elucidated. The present study investigates whether HPV infection is correlated with changes in plasmatic arginase activity and cervical ARG1 and ARG2 mRNA expression among infected women negative for intraepithelial lesions (NIL). MATERIEL AND METHODS The present study included 300 women. The plasmatic arginase activity was evaluated by a colorimetric assay. Cervical HPV was detected by real-time PCR. The circulating viral load and ARG1 and ARG2 mRNA expression quantification were performed by quantitative real-time PCR. RESULTS A significant increase in plasma arginase activity and ARG1 and ARG2 mRNA expression levels in cervical cells was observed among HPV-positive women compared to the HPV-negative group. The highest levels were significantly associated with oncogenic HPV, and increased arginase activity was associated with a high HPV circulating viral load. Moreover, the highest levels of arginase activity were observed in oncogenic HPV-positive inflammatory smears. DISCUSSION These data suggest that HPV could modulate arginase activity and expression, which may restrict arginine bioavailability and inhibit this amino acid's antiviral properties. CONCLUSION Our findings revealed that arginase activity and isoform gene expression were upregulated in women with HPV infection, particularly the oncogenic HPV types.
Collapse
Affiliation(s)
- Moufida Souid
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Higher Institute of Biotechnology of Monastir, Monastir University, Monastir, Tunisia
| | - Jaweher Bday
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Higher Institute of Biotechnology of Monastir, Monastir University, Monastir, Tunisia
| | - Sameh Souissi
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Higher Institute of Biotechnology of Monastir, Monastir University, Monastir, Tunisia
| | - Randa Ghedira
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
| | - Sallouha Gabbouj
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
| | | | - Dhekra Toumi
- Department of Gynecology and Obstetrics, University Hospital of Monastir, Monastir, Tunisia
| | - Haifa Bergaoui
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Department of Gynecology and Obstetrics, University Hospital of Monastir, Monastir, Tunisia
| | - Ines Zouari
- Department of Gynecology and Obstetrics, University Hospital of Monastir, Monastir, Tunisia
| | - Raja Faleh
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Department of Gynecology and Obstetrics, University Hospital of Monastir, Monastir, Tunisia
| | - Abdelfatteh Zakhama
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
| | - Elham Hassen
- Molecular Immuno-Oncology Laboratory, Monastir University, Monastir, Tunisia
- Higher Institute of Biotechnology of Monastir, Monastir University, Monastir, Tunisia
| |
Collapse
|
4
|
Xu Z, Liu Y, Ma R, Chen J, Qiu J, Du S, Li C, Wu Z, Yang X, Chen Z, Chen T. Thermosensitive Hydrogel Incorporating Prussian Blue Nanoparticles Promotes Diabetic Wound Healing via ROS Scavenging and Mitochondrial Function Restoration. ACS APPLIED MATERIALS & INTERFACES 2022; 14:14059-14071. [PMID: 35298140 DOI: 10.1021/acsami.1c24569] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Diabetic foot ulcer is a serious complication in diabetes patients, imposing a serious physical and economic burden to patients and to the healthcare system as a whole. Oxidative stress is thought to be a key driver of the pathogenesis of such ulcers. However, no antioxidant drugs have received clinical approval to date, underscoring the need for the further development of such medications. Hydrogels can be applied directly to the wound site, wherein they function to prevent infection and maintain local moisture concentrations, in addition to serving as a reservoir for the delivery of a range of therapeutic compounds with the potential to expedite wound healing in a synergistic manner. Herein, we synthesized Prussian blue nanoparticles (PBNPs) capable of efficiently scavenging reactive oxygen species (ROS) owing to their ability to mimic the activity of catalase (CAT), peroxidase (POD), and superoxide dismutase (SOD). In the context of in vitro oxidative stress, these PBNPs were able to protect against cytotoxicity, protect mitochondria from oxidative stress-related damage, and restore nuclear factor erythroid 2-related factor 2 (NRF2)/heme oxygenase-1 (HO-1) pathway activity. To expand on these results in an in vivo context, we prepared a thermosensitive poly (d,l-lactide)-poly(ethylene glycol)-poly(d,l-lactide) (PDLLA-PEG-PDLLA) hydrogel (PLEL)-based wound dressing in which PBNPs had been homogenously incorporated, and we then used this dressing as a platform for controlled PBNP release. The resultant PBNPs@PLEL wound dressing was able to improve diabetic wound healing, decrease ROS production, promote angiogenesis, and reduce pro-inflammatory interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) levels within diabetic wounds. Overall, our results suggest that this PBNPs@PLEL platform holds great promise as a treatment for diabetic foot ulcers.
Collapse
Affiliation(s)
- Zhao Xu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yujing Liu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Rui Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jinmei Qiu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Shuang Du
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Chengcheng Li
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zihan Wu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaofan Yang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Tongkai Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| |
Collapse
|
5
|
Souid M, Ghedira R, Souissi S, Bouzgarrou N, Gabbouj S, Shini-Hadhri S, Rhim MS, Boukadida A, Toumi D, Faleh R, Bouaouina N, Zakhama A, Hassen E. Arginase is involved in cervical lesions progression and severity. Immunobiology 2022; 227:152189. [DOI: 10.1016/j.imbio.2022.152189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 02/03/2022] [Accepted: 02/12/2022] [Indexed: 11/15/2022]
|
6
|
Shosha E, Fouda AY, Lemtalsi T, Haigh S, Fulton D, Ibrahim A, Al-Shabrawey M, Caldwell RW, Caldwell RB. Endothelial arginase 2 mediates retinal ischemia/reperfusion injury by inducing mitochondrial dysfunction. Mol Metab 2021; 53:101273. [PMID: 34139341 PMCID: PMC8274341 DOI: 10.1016/j.molmet.2021.101273] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/05/2021] [Accepted: 06/11/2021] [Indexed: 12/19/2022] Open
Abstract
Objective Retinal ischemic disease is a major cause of vision loss. Current treatment options are limited to late-stage diseases, and the molecular mechanisms of the initial insult are not fully understood. We have previously shown that the deletion of the mitochondrial arginase isoform, arginase 2 (A2), limits neurovascular injury in models of ischemic retinopathy. Here, we investigated the involvement of A2-mediated alterations in mitochondrial dynamics and function in the pathology. Methods We used wild-type (WT), global A2 knockout (A2KO-) mice, cell-specific A2 knockout mice subjected to retinal ischemia/reperfusion (I/R), and bovine retinal endothelial cells (BRECs) subjected to an oxygen-glucose deprivation/reperfusion (OGD/R) insult. We used western blotting to measure levels of cell stress and death markers and the mitochondrial fragmentation protein, dynamin related protein 1 (Drp1). We also used live cell mitochondrial labeling and Seahorse XF analysis to evaluate mitochondrial fragmentation and function, respectively. Results We found that the global deletion of A2 limited the I/R-induced disruption of retinal layers, fundus abnormalities, and albumin extravasation. The specific deletion of A2 in endothelial cells was protective against I/R-induced neurodegeneration. The OGD/R insult in BRECs increased A2 expression and induced cell stress and cell death, along with decreased mitochondrial respiration, increased Drp1 expression, and mitochondrial fragmentation. The overexpression of A2 in BREC also decreased mitochondrial respiration, promoted increases in the expression of Drp1, mitochondrial fragmentation, and cell stress and resulted in decreased cell survival. In contrast, the overexpression of the cytosolic isoform, arginase 1 (A1), did not affect these parameters. Conclusions This study is the first to show that A2 in endothelial cells mediates retinal ischemic injury through a mechanism involving alterations in mitochondrial dynamics and function. Ischemic retinopathy is a common feature of blinding eye disease. Arginase 2 overexpression in endothelial cells induces mitochondrial dysfunction. Endothelial-specific arginase 2 deletion improves neuronal survival after ischemia. Endothelial cell arginase 2 plays a crucial role in ischemic retinal injury.
Collapse
Affiliation(s)
- Esraa Shosha
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Vision Discovery Institute, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Abdelrahman Y Fouda
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Department of Clinical Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Vision Discovery Institute, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Tahira Lemtalsi
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Vision Discovery Institute, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Stephen Haigh
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - David Fulton
- Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Ahmed Ibrahim
- Vision Discovery Institute, Augusta University, Augusta, GA, USA; Wayne State University, Department of Ophthalmology, Visual, and Anatomical Sciences, Department of Pharmacology, Detroit, MI, USA; Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Mohamed Al-Shabrawey
- Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Oral Biology, Dental College of Georgia, Augusta, GA, USA
| | - R William Caldwell
- Vision Discovery Institute, Augusta University, Augusta, GA, USA; Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Vision Discovery Institute, Augusta University, Augusta, GA, USA; Charlie Norwood VA Medical Center, Augusta, GA, USA.
| |
Collapse
|
7
|
Polis B, Karasik D, Samson AO. Alzheimer's disease as a chronic maladaptive polyamine stress response. Aging (Albany NY) 2021; 13:10770-10795. [PMID: 33811757 PMCID: PMC8064158 DOI: 10.18632/aging.202928] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 03/27/2021] [Indexed: 12/21/2022]
Abstract
Polyamines are nitrogen-rich polycationic ubiquitous bioactive molecules with diverse evolutionary-conserved functions. Their activity interferes with numerous genes' expression resulting in cell proliferation and signaling modulation. The intracellular levels of polyamines are precisely controlled by an evolutionary-conserved machinery. Their transient synthesis is induced by heat stress, radiation, and other traumatic stimuli in a process termed the polyamine stress response (PSR). Notably, polyamine levels decline gradually with age; and external supplementation improves lifespan in model organisms. This corresponds to cytoprotective and reactive oxygen species scavenging properties of polyamines. Paradoxically, age-associated neurodegenerative disorders are characterized by upsurge in polyamines levels, indicating polyamine pleiotropic, adaptive, and pathogenic roles. Specifically, arginase overactivation and arginine brain deprivation have been shown to play an important role in Alzheimer's disease (AD) pathogenesis. Here, we assert that a universal short-term PSR associated with acute stimuli is beneficial for survival. However, it becomes detrimental and maladaptive following chronic noxious stimuli, especially in an aging organism. Furthermore, we regard cellular senescence as an adaptive response to stress and suggest that PSR plays a central role in age-related neurodegenerative diseases' pathogenesis. Our perspective on AD proposes an inclusive reassessment of the causal relationships between the classical hallmarks and clinical manifestation. Consequently, we offer a novel treatment strategy predicated upon this view and suggest fine-tuning of arginase activity with natural inhibitors to preclude or halt the development of AD-related dementia.
Collapse
Affiliation(s)
- Baruh Polis
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - David Karasik
- Hebrew SeniorLife, Hinda and Arthur Marcus Institute for Aging Research, Boston, MA 02131, USA
- Musculoskeletal Genetics Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| | - Abraham O. Samson
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| |
Collapse
|
8
|
Chen S, Chen H, Zhong Y, Ge Y, Li C, Qiao Z, Zhu J. Insulin-like growth factor-binding protein 3 inhibits angiotensin II-induced aortic smooth muscle cell phenotypic switch and matrix metalloproteinase expression. Exp Physiol 2020; 105:1827-1839. [PMID: 32936966 DOI: 10.1113/ep088927] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/15/2020] [Indexed: 12/28/2022]
Abstract
NEW FINDINGS What is the central question of this study? Insulin-like growth factor 1 and its major binding protein insulin-like growth factor binding protein 3 (IGFBP3) are involved in collagen deregulation in several cardiovascular diseases: what is the role of IGFBP3 in thoracic aortic dissection and does it regulate aortic smooth muscle cells' phenotypic switch? What is the main finding and its importance? IGFBP3 inhibits aortic smooth muscle cells' phenotypic switch from a contractile to a synthetic phenotype, decreases matrix metalloproteinase 9 activation and suppresses elastin degradation. These findings provide a better understanding of the pathogenesis of thoracic aortic dissection. ABSTRACT Thoracic aortic dissection (TAD) is characterized by aortic media degeneration and is a highly lethal disease. An aortic smooth muscle cell (AoSMC) phenotypic switch is considered a key pathophysiological change in TAD. Insulin-like growth factor binding protein 3 (IGFBP3) was found to be downregulated in aortic tissues of TAD patients. The present work aimed to study the function of IGFBP3 in AoSMCs' phenotypic switch and matrix metalloproteinase (MMP) expression. We established a mouse model of TAD by angiotensin (Ang) II infusion to β-aminopropionitrile-administrated mice, and found decreased IGFBP3 expression accompanied by aortic dilatation and elastin degradation in vivo. Further, mouse (m)AoSMCs were isolated from mouse thoracic aorta and treated with Ang II. Ang II induced downregulation of IGFBP3 in vitro. To further study the function of IGFBP3, primary mAoSMCs were infected with adenovirus expressing IGFBP3 followed by Ang II induction. Enforced upregulation of IGFBP3 decreased MMP9 expression and activation as well as increasing tissue inhibitor of metalloproteinase (TIMP) 1 expression in Ang II-induced mAoSMCs. No difference was observed in MMP2 and TIMP3 expression. IGFBP3 suppressed subsequent Ang II-induced elastin degradation in vitro. IGFBP3 inhibited Ang II-induced mAoSMCs' phenotypic switch as evidenced by increased smooth muscle actin α-2 (ACTA2) and myosin heavy chain 11 (MYH11) expression and decreased secreted phosphoprotein 1 (SPP1) and vimentin expression. Taken together, the present study demonstrates the role of IGFBP3 in preserving AoSMCs' contractile state and reducing MMP9 activation and thus promoting elastic fibre synthesis, which provides a better understanding of the pathogenesis of TAD.
Collapse
Affiliation(s)
- Suwei Chen
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Hong Chen
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yongliang Zhong
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yipeng Ge
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chengnan Li
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Zhiyu Qiao
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Junming Zhu
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
9
|
Koo BH, Won MH, Kim YM, Ryoo S. p32-Dependent p38 MAPK Activation by Arginase II Downregulation Contributes to Endothelial Nitric Oxide Synthase Activation in HUVECs. Cells 2020; 9:cells9020392. [PMID: 32046324 PMCID: PMC7072651 DOI: 10.3390/cells9020392] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/26/2020] [Accepted: 02/05/2020] [Indexed: 12/30/2022] Open
Abstract
Arginase II reciprocally regulates endothelial nitric oxide synthase (eNOS) through a p32-dependent Ca2+ control. We investigated the signaling pathway of arginase II-dependent eNOS phosphorylation. Western blot analysis was applied for examining protein activation and [Ca2+]c was analyzed by microscopic and FACS analyses. Nitric oxide (NO) and reactive oxygen species (ROS) productions were measured using specific fluorescent dyes under microscopy. NO signaling pathway was tested by measuring vascular tension. Following arginase II downregulation by chemical inhibition or gene knockout (KO, ArgII−/−), increased eNOS phosphorylation at Ser1177 and decreased phosphorylation at Thr495 was depend on p38 MAPK activation, which induced by CaMKII activation through p32-dependent increase in [Ca2+]c. The protein amount of p32 negatively regulated p38 MAPK activation. p38 MAPK contributed to Akt-induced eNOS phosphorylation at Ser1177 that resulted in accelerated NO production and reduced reactive oxygen species production in aortic endothelia. In vascular tension assay, p38 MAPK inhibitor decreased acetylcholine-induced vasorelaxation responses and increased phenylephrine-dependent vasoconstrictive responses. In ApoE−/− mice fed a high cholesterol diet, arginase II inhibition restored p32/CaMKII/p38 MAPK/Akt/eNOS signaling cascade that was attenuated by p38 MAPK inhibition. Here, we demonstrated a novel signaling pathway contributing to understanding of the relationship between arginase II, endothelial dysfunction, and atherogenesis.
Collapse
Affiliation(s)
- Bon-Hyeock Koo
- Department of Biological Sciences, Kangwon National University, Chuncheon, Gangwon 24341, Korea;
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea;
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Korea;
| | - Sungwoo Ryoo
- Department of Biological Sciences, Kangwon National University, Chuncheon, Gangwon 24341, Korea;
- Correspondence: ; Tel.: +82-33-250-8534; Fax: +82-33-251-3990
| |
Collapse
|
10
|
Is the Arginase Pathway a Novel Therapeutic Avenue for Diabetic Retinopathy? J Clin Med 2020; 9:jcm9020425. [PMID: 32033258 PMCID: PMC7073619 DOI: 10.3390/jcm9020425] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 01/31/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetic retinopathy (DR) is the leading cause of blindness in working age Americans. Clinicians diagnose DR based on its characteristic vascular pathology, which is evident upon clinical exam. However, extensive research has shown that diabetes causes significant neurovascular dysfunction prior to the development of clinically apparent vascular damage. While laser photocoagulation and/or anti-vascular endothelial growth factor (VEGF) therapies are often effective for limiting the late-stage vascular pathology, we still do not have an effective treatment to limit the neurovascular dysfunction or promote repair during the early stages of DR. This review addresses the role of arginase as a mediator of retinal neurovascular injury and therapeutic target for early stage DR. Arginase is the ureohydrolase enzyme that catalyzes the production of L-ornithine and urea from L-arginine. Arginase upregulation has been associated with inflammation, oxidative stress, and peripheral vascular dysfunction in models of both types of diabetes. The arginase enzyme has been identified as a therapeutic target in cardiovascular disease and central nervous system disease including stroke and ischemic retinopathies. Here, we discuss and review the literature on arginase-induced retinal neurovascular dysfunction in models of DR. We also speculate on the therapeutic potential of arginase in DR and its related underlying mechanisms.
Collapse
|
11
|
Koo BH, Hwang HM, Yi BG, Lim HK, Jeon BH, Hoe KL, Kwon YG, Won MH, Kim YM, Berkowitz DE, Ryoo S. Arginase II Contributes to the Ca 2+/CaMKII/eNOS Axis by Regulating Ca 2+ Concentration Between the Cytosol and Mitochondria in a p32-Dependent Manner. J Am Heart Assoc 2019; 7:e009579. [PMID: 30371203 PMCID: PMC6222941 DOI: 10.1161/jaha.118.009579] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Arginase II activity contributes to reciprocal regulation of endothelial nitric oxide synthase (eNOS). We tested the hypotheses that arginase II activity participates in the regulation of Ca2+/Ca2+/calmodulin‐dependent kinase II/eNOS activation, and this process is dependent on mitochondrial p32. Methods and Results Downregulation of arginase II increased the concentration of cytosolic Ca2+ ([Ca2+]c) and decreased mitochondrial Ca2+ ([Ca2+]m) in microscopic and fluorescence‐activated cell sorting analyses, resulting in augmented eNOS Ser1177 phosphorylation and decreased eNOS Thr495 phosphorylation through Ca2+/Ca2+/calmodulin‐dependent kinase II. These changes were observed in human umbilical vein endothelial cells treated with small interfering RNA against p32 (sip32). Using matrix‐assisted laser desorption/ionization time‐of‐flight mass spectrometry, fluorescence immunoassay, and ion chromatography, inhibition of arginase II reduced the amount of spermine, a binding molecule, and the release of Ca2+ from p32. In addition, arginase II gene knockdown using small interfering RNA and knockout arginase II‐null mice resulted in reduced p32 protein level. In the aortas of wild‐type mice, small interfering RNA against p32 induced eNOS Ser1177 phosphorylation and enhanced NO‐dependent vasorelaxation. Arginase activity, p32 protein expression, spermine amount, and [Ca2+]m were increased in the aortas from apolipoprotein E (ApoE−/−) mice fed a high‐cholesterol diet, and intravenous administration of small interfering RNA against p32 restored Ca2+/Ca2+/calmodulin‐dependent kinase II‐dependent eNOS Ser1177 phosphorylation and improved endothelial dysfunction. The effects of arginase II downregulation were not associated with elevated NO production when tested in aortic endothelia from eNOS knockout mice. Conclusions These data demonstrate a novel function of arginase II in regulation of Ca2+‐dependent eNOS phosphorylation. This novel mechanism drives arginase activation, mitochondrial dysfunction, endothelial dysfunction, and atherogenesis.
Collapse
Affiliation(s)
- Bon-Hyeock Koo
- 1 Department of Biology School of medicine Kangwon National University Chuncheon Korea
| | - Hye-Mi Hwang
- 1 Department of Biology School of medicine Kangwon National University Chuncheon Korea
| | - Bong-Gu Yi
- 1 Department of Biology School of medicine Kangwon National University Chuncheon Korea
| | - Hyun Kyo Lim
- 4 Department of Anesthesiology and Pain Medicine Yonsei University Wonju College of Medicine Wonju Korea
| | - Byeong Hwa Jeon
- 5 Infectious Signaling Network Research Center Department of Physiology School of Medicine Chungnam National University Daejeon Korea
| | - Kwang Lae Hoe
- 6 Department of New Drug Discovery and Development Chungnam National University Daejeon Korea
| | | | - Moo-Ho Won
- 2 Department of Neurobiology School of medicine Kangwon National University Chuncheon Korea
| | - Young Myeong Kim
- 3 College of Natural Sciences and Departments of Molecular and Cellular Biochemistry School of medicine Kangwon National University Chuncheon Korea
| | - Dan E Berkowitz
- 8 Department of Anesthesiology and Critical Care Medicine Johns Hopkins University Baltimore MD
| | - Sungwoo Ryoo
- 1 Department of Biology School of medicine Kangwon National University Chuncheon Korea
| |
Collapse
|
12
|
Arginase II activity regulates cytosolic Ca 2+ level in a p32-dependent manner that contributes to Ca 2+-dependent vasoconstriction in native low-density lipoprotein-stimulated vascular smooth muscle cells. Exp Mol Med 2019; 51:1-12. [PMID: 31155612 PMCID: PMC6545325 DOI: 10.1038/s12276-019-0262-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/19/2019] [Accepted: 02/26/2019] [Indexed: 12/04/2022] Open
Abstract
Although arginase II (ArgII) is abundant in mitochondria, Ca2+-accumulating organelles, the relationship between ArgII activity and Ca2+ translocation into mitochondria and the regulation of cytosolic Ca2+ signaling are completely unknown. We investigated the effects of ArgII activity on mitochondrial Ca2+ uptake through mitochondrial p32 protein (p32m) and on CaMKII-dependent vascular smooth muscle cell (VSMC) contraction. Native low-density lipoprotein stimulation induced an increase in [Ca2+]m as measured by CoCl2-quenched calcein-AM fluorescence, which was prevented by Arg inhibition in hAoSMCs and reduced in mAoSMCs from ArgII−/− mice. Conversely, [Ca2+]c analyzed with Fluo-4 AM was increased by Arg inhibition and ArgII gene knockout. The increased [Ca2+]c resulted in CaMKII and MLC 20 phosphorylation, which was associated with enhanced vasoconstriction activity to phenylephrine (PE) in the vascular tension assay. Cy5-tagged siRNA against mitochondrial p32 mRNA (sip32m) abolished mitochondrial Ca2+ uptake and induced activation of CaMKII. Spermine, a polyamine, induced mitochondrial Ca2+ uptake and dephosphorylation of CaMKII and was completely inhibited by sip32m incubation. In mAoSMCs from ApoE-null mice fed a high-cholesterol diet (ApoE−/− +HCD), Arg activity was increased, and spermine concentration was higher than that of wild-type mice. Furthermore, [Ca2+]m and p32m levels were elevated, and CaMKII phosphorylation was reduced in mAoSMCs from ApoE−/− +HCD. In vascular tension experiments, an attenuated response to vasoconstrictors in de-endothelialized aorta from ApoE−/− +HCD was recovered by incubation of sip32m. ArgII activity-dependent production of spermine augments Ca2+ transition from the cytosol to the mitochondria in a p32m-dependent manner and regulates CaMKII-dependent constriction in VSMCs. Researchers have illuminated how a protein, arginase II (ArgII), is involved in development of vascular diseases such as atherosclerosis, or narrowing of the arteries by plaque deposits. Blood vessel diameter is regulated by layers of muscle; the balance between constriction and relaxation is critical for blood flow and vascular health. Increased ArgII is known to be a factor in arterial disease; however, the details of regulation, and how they relate to plaque deposition, remain poorly understood. Sungwoo Ryoo at Kangwon National University, Chuncheon, South Korea and co-workers investigated how ArgII levels affect arterial constriction and relaxation in mice. Decreasing ArgII restored the muscle cells’ contraction response by preventing excessive calcium accumulation in the cellular powerhouse, mitochondria. These results may aid in developing treatments for one of the leading causes of death worldwide.
Collapse
|
13
|
Krystofova J, Pathipati P, Russ J, Sheldon A, Ferriero D. The Arginase Pathway in Neonatal Brain Hypoxia-Ischemia. Dev Neurosci 2019; 40:437-450. [PMID: 30995639 PMCID: PMC6784534 DOI: 10.1159/000496467] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/20/2018] [Indexed: 12/11/2022] Open
Abstract
Brain damage after hypoxia-ischemia (HI) occurs in an age-dependent manner. Neuroprotective strategies assumed to be effective in adults might have deleterious effects in the immature brain. In order to create effective therapies, the complex pathophysiology of HI in the developing brain requires exploring new mechanisms. Critical determinants of neuronal survival after HI are the extent of vascular dysfunction, inflammation, and oxidative stress, followed later by tissue repair. The key enzyme of these processes in the human body is arginase (ARG) that acts via the bioavailability of nitric oxide, and the synthesis of polyamines and proline. ARG is expressed throughout the brain in different cells. However, little is known about the effect of ARG in pathophysiological states of the brain, especially hypoxia-ischemia. Here, we summarize the role of ARG during neurodevelopment as well as in various brain pathologies.
Collapse
Affiliation(s)
- Jana Krystofova
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA,
| | - Praneeti Pathipati
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Jeffrey Russ
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Ann Sheldon
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| | - Donna Ferriero
- Department of Pediatrics, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
14
|
Choi CI, Koo BH, Hong D, Kwon HJ, Hoe KL, Won MH, Kim YM, Lim HK, Ryoo S. Resveratrol is an arginase inhibitor contributing to vascular smooth muscle cell vasoconstriction via increasing cytosolic calcium. Mol Med Rep 2019; 19:3767-3774. [PMID: 30896798 DOI: 10.3892/mmr.2019.10035] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/28/2019] [Indexed: 11/05/2022] Open
Abstract
The contractility of vascular smooth muscle cells (VSMCs) controls the lumen diameter of vessels, thus serving a role in regulating blood pressure and organ blood flow. Although arginases are known to have numerous effects in the biological activities of VSMCs, the effects of arginase II on the constriction of VSMCs has not yet been investigated. When conducting a natural products screen for an inhibitor against arginase, the present study identified that a relatively high concentration of resveratrol (RSV) exhibited arginase inhibitory activity. Therefore, the present study investigated whether RSV could regulate VSMCs contractions and the underlying mechanism. Arginase inhibition by RSV led to an increase in the concentration of the substrate L‑Arg and an accompanying increase in the cytosol Ca2+ concentration [(Ca2+)c] in VSMCs. The increased [Ca2+]c induced by RSV and L‑Arg treatments resulted in CaMKII‑dependent MLC20 phosphorylation. The effects of RSV on VSMCs were maintained even when VSMCs were pre‑treated with sirtinol, an inhibitor of Sirt proteins. In a vascular tension assay with de‑endothelialized aortic vessels, vasoconstrictor responses, which were measured using phenylephrine (PE), were significantly enhanced in the RSV‑ and L‑Arg‑treated vessels. Therefore, although arginase inhibition has exhibited beneficial effects in various diseases, care is required when considering administration of an arginase inhibitor to patients with vessels endothelial dysfunction as RSV can induce vessel contraction.
Collapse
Affiliation(s)
- Chang Ik Choi
- Department of Anesthesiology and Pain Medicine, Yonsei University Wonju College of Medicine, Wonju, Gangwon 26426, Republic of Korea
| | - Bon Hyeock Koo
- Department of Biology, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Dongeui Hong
- Department of Anesthesiology and Pain Medicine, Yonsei University Wonju College of Medicine, Wonju, Gangwon 26426, Republic of Korea
| | - Hyung Joo Kwon
- Department of Microbiology, School of Medicine, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Kwang Lae Hoe
- New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Moo Ho Won
- Department of Neurobiology, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Young Myeong Kim
- Department of Molecular and Cellular Biochemistry, and Neurobiology, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Hyun Kyo Lim
- Department of Anesthesiology and Pain Medicine, Yonsei University Wonju College of Medicine, Wonju, Gangwon 26426, Republic of Korea
| | - Sungwoo Ryoo
- Department of Biology, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| |
Collapse
|