1
|
Zhang L, Yang J, Liu W, Ding Q, Sun S, Zhang S, Wang N, Wang Y, Xi S, Liu C, Ding C, Li C. A phellinus igniarius polysaccharide/chitosan-arginine hydrogel for promoting diabetic wound healing. Int J Biol Macromol 2023; 249:126014. [PMID: 37517765 DOI: 10.1016/j.ijbiomac.2023.126014] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/01/2023]
Abstract
Inadequate angiogenesis and inflammation at the wound site have always been a major threat to skin wounds, especially for diabetic wounds that are difficult to heal. Therefore, hydrogel dressings with angiogenesis and antibacterial properties are very necessary in practical applications. This study reported a hydrogel (PCA) based on L-arginine conjugated chitosan (CA) and aldehyde functionalized polysaccharides of Phellinus igniarius (OPPI) as an antibacterial and pro-angiogenesis dressing for wound repair in diabetes for the first time. and discussed its possible mechanism for promoting wound healing. The results showed that PCA had good antioxidant, antibacterial, biological safety and other characteristics, and effectively promoted the healing course of diabetic wound model. In detail, the H&E and Masson staining results showed that PCA promoted normal epithelial formation and collagen deposition. The Western blot results confirmed that PCA decreased the inflammation by inhibiting the IKBα/NF-κB signaling pathway and enhanced angiogenesis by adjusting the level of HIF-1α. In conclusion, PCA is a promising candidate for promoting wound healing in diabetes. Graphic abstract.
Collapse
Affiliation(s)
- Lifeng Zhang
- Engineering Research Center of the Ministry of Education, Jilin Agricultural University, Changchun 130118, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Jiali Yang
- Engineering Research Center of the Ministry of Education, Jilin Agricultural University, Changchun 130118, China; College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Wencong Liu
- School of Food and Pharmaceutical Engineering, Wuzhou University, Wuzhou 543002, China
| | - Qiteng Ding
- College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Shuwen Sun
- College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Shuai Zhang
- College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Ning Wang
- College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Yue Wang
- College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Siyu Xi
- College of traditional Chinese Medicine, Jilin Agricultural University, Changchun 130118, China
| | - Chunyu Liu
- Engineering Research Center of the Ministry of Education, Jilin Agricultural University, Changchun 130118, China
| | - Chuanbo Ding
- College of traditional Chinese Medicine, Jilin Agriculture Science and Technology College, Jilin 132101, China.
| | - Changtian Li
- Engineering Research Center of the Ministry of Education, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
2
|
Harris ZM, Sun Y, Joerns J, Clark B, Hu B, Korde A, Sharma L, Shin HJ, Manning EP, Placek L, Unutmaz D, Stanley G, Chun H, Sauler M, Rajagopalan G, Zhang X, Kang MJ, Koff JL. Epidermal Growth Factor Receptor Inhibition Is Protective in Hyperoxia-Induced Lung Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9518592. [PMID: 36193076 PMCID: PMC9526641 DOI: 10.1155/2022/9518592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 01/01/2023]
Abstract
Aims Studies have linked severe hyperoxia, or prolonged exposure to very high oxygen levels, with worse clinical outcomes. This study investigated the role of epidermal growth factor receptor (EGFR) in hyperoxia-induced lung injury at very high oxygen levels (>95%). Results Effects of severe hyperoxia (100% oxygen) were studied in mice with genetically inhibited EGFR and wild-type littermates. Despite the established role of EGFR in lung repair, EGFR inhibition led to improved survival and reduced acute lung injury, which prompted an investigation into this protective mechanism. Endothelial EGFR genetic knockout did not confer protection. EGFR inhibition led to decreased levels of cleaved caspase-3 and poly (ADP-ribosyl) polymerase (PARP) and decreased terminal dUTP nick end labeling- (TUNEL-) positive staining in alveolar epithelial cells and reduced ERK activation, which suggested reduced apoptosis in vivo. EGFR inhibition decreased hyperoxia (95%)-induced apoptosis and ERK in murine alveolar epithelial cells in vitro, and CRISPR-mediated EGFR deletion reduced hyperoxia-induced apoptosis and ERK in human alveolar epithelial cells in vitro. Innovation. This work defines a protective role of EGFR inhibition to decrease apoptosis in lung injury induced by 100% oxygen. This further characterizes the complex role of EGFR in acute lung injury and outlines a novel hyperoxia-induced cell death pathway that warrants further study. Conclusion In conditions of severe hyperoxia (>95% for >24 h), EGFR inhibition led to improved survival, decreased lung injury, and reduced cell death. These findings further elucidate the complex role of EGFR in acute lung injury.
Collapse
Affiliation(s)
- Zachary M. Harris
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Ying Sun
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - John Joerns
- Division of Pulmonary and Critical Care; Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA 75390
| | - Brian Clark
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Buqu Hu
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Asawari Korde
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Lokesh Sharma
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Hyeon Jun Shin
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Edward P. Manning
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
- VA Connecticut Healthcare System, West Haven, CT, USA
| | - Lindsey Placek
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA
| | - Derya Unutmaz
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut 06032, USA
| | - Gail Stanley
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Hyung Chun
- Section of Cardiovascular Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Maor Sauler
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Govindarajan Rajagopalan
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Xuchen Zhang
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Min-Jong Kang
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| | - Jonathan L. Koff
- Section of Pulmonary, Critical Care, and Sleep Medicine; Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA 06510
| |
Collapse
|
3
|
Integrated Metabolomics and Transcriptomic Analysis of Hepatopancreas in Different Living Status Macrobrachium nipponense in Response to Hypoxia. Antioxidants (Basel) 2021; 11:antiox11010036. [PMID: 35052540 PMCID: PMC8772856 DOI: 10.3390/antiox11010036] [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: 11/23/2021] [Revised: 12/19/2021] [Accepted: 12/22/2021] [Indexed: 12/16/2022] Open
Abstract
As the basic element of aerobic animal life, oxygen participates in most physiological activities of animals. Hypoxia stress is often the subject of aquatic animal research. Macrobrachium nipponense, an economically important aquatic animal in southern China, has been affected by hypoxia for many years and this has resulted in a large amount of economic loss due to its sensitivity to hypoxia; Metabolism and transcriptome data were combined in the analysis of the hepatopancreas of M. nipponense in different physiological states under hypoxia; A total of 108, 86, and 48 differentially expressed metabolites (DEMs) were found in three different comparisons (survived, moribund, and dead shrimps), respectively. Thirty-two common DEMs were found by comparing the different physiological states of M. nipponense with the control group in response to hypoxia. Twelve hypoxia-related genes were identified by screening and analyzing common DEMs. GTP phosphoenolpyruvate carboxykinase (PEPCK) was the only differentially expressed gene that ranked highly in transcriptome analysis combined with metabolome analysis. PEPCK ranked highly both in transcriptome analysis and in combination with metabolism analysis; therefore, it was considered to have an important role in hypoxic response. This manuscript fills the one-sidedness of the gap in hypoxia transcriptome analysis and reversely deduces several new genes related to hypoxia from metabolites. This study contributes to the clarification of the molecular process associated with M. nipponense under hypoxic stress.
Collapse
|
4
|
Garate-Carrillo A, Navarrete-Yañez V, Ortiz-Vilchis P, Guevara G, Castillo C, Mendoza-Lorenzo P, Ceballos G, Ortiz-Flores M, Najera N, Bustamante-Pozo MM, Rubio-Gayosso I, Villarreal F, Ramirez-Sanchez I. Arginase inhibition by (-)-Epicatechin reverses endothelial cell aging. Eur J Pharmacol 2020; 885:173442. [PMID: 32795514 PMCID: PMC7418791 DOI: 10.1016/j.ejphar.2020.173442] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 12/16/2022]
Abstract
Endothelial dysfunction (EnD) occurs with aging and endothelial nitric oxide (NO) production by NO synthase (NOS) can be impaired. Low NO levels have been linked to increased arginase (Ar) activity as Ar competes with NOS for L-arginine. The inhibition of Ar activity can reverse EnD and (-)-epicatechin (Epi) inhibits myocardial Ar activity. In this study, through in silico modeling we demonstrate that Epi interacts with Ar similarly to its inhibitor Norvaline (Norv). Using in vitro and in vivo models of aging, we examined Epi and Norv-inhibition of Ar activity and its endothelium-protective effects. Bovine coronary artery endothelial cells (BCAEC) were treated with Norv (10 μM), Epi (1 μM) or the combination (Epi + Norv) for 48 h. Ar activity increased in aged BCAEC, with decreased NO generation. Treatment decreased Ar activity to levels seen in young cells. Epi and Epi + Norv decreased nitrosylated Ar levels by ~25% in aged cells with lower oxidative stress (~25%) (dihydroethidium) levels. In aged cells, Epi and Epi + Norv restored the eNOS monomer/dimer ratio, protein expression levels and NO production to those of young cells. Furthermore, using 18 month old rats 15 days of treatment with either Epi (1 mg/kg), Norv (10 mg/kg) or combo, decreased hypertension and improved aorta vasorelaxation to acetylcholine, blood NO levels and tetra/dihydribiopterin ratios in cultured rat aortic endothelial cells. In conclusion, results provide evidence that inhibiting Ar with Epi reverses aged-related loss of eNOS function and improves vascular function through the modulation of Ar and eNOS protein levels and activity.
Collapse
Affiliation(s)
- Alejandra Garate-Carrillo
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | - Viridiana Navarrete-Yañez
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | - Pilar Ortiz-Vilchis
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | - Gustavo Guevara
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | - Carmen Castillo
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | - Patricia Mendoza-Lorenzo
- División Académica de Ciencias Básicas, Unidad Chontalpa, Universidad Juárez Autónoma de Tabasco, Tabasco, Mexico
| | - Guillermo Ceballos
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | - Miguel Ortiz-Flores
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | - Nayelli Najera
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | - Moises Muratt Bustamante-Pozo
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | - Ivan Rubio-Gayosso
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico
| | - Francisco Villarreal
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA; VA San Diego Health Care, San Diego, CA, USA
| | - Israel Ramirez-Sanchez
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico.
| |
Collapse
|
5
|
Trittmann JK, Almazroue H, Jin Y, Nelin LD. DDAH1 regulates apoptosis and angiogenesis in human fetal pulmonary microvascular endothelial cells. Physiol Rep 2020; 7:e14150. [PMID: 31209995 PMCID: PMC6579941 DOI: 10.14814/phy2.14150] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 05/30/2019] [Accepted: 06/01/2019] [Indexed: 01/17/2023] Open
Abstract
Nitric Oxide (NO) is an endogenous pulmonary vasodilator produced by endothelial NO synthase (eNOS). Asymmetric dimethyl L‐arginine (ADMA) is an endogenous inhibitor of eNOS activity. In endothelial cells, ADMA is hydrolyzed to L‐citrulline primarily by dimethylarginine dimethyl‐aminohydrolase‐1 (DDAH1). We tested the hypothesis that DDAH1 expression is essential for maintaining NO production in human fetal pulmonary microvascular endothelial cells (hfPMVEC), such that knockdown of DDAH1 expression will lead to decreased NO production resulting in less caspase‐3 activation and less tube formation. We found that hfPMVEC transfected with DDAH1 siRNA had lower NO production than control, with no difference in eNOS protein levels between groups. hfPMVEC transfected with DDAH1 siRNA had lower protein levels of cleaved caspase‐3 and ‐8 than control. Both DDAH1 siRNA‐ and ADMA‐treated hfPMVEC had greater numbers of viable cells than controls. Angiogenesis was assessed using tube formation assays in matrigel, and tube formation was lower after either DDAH1 siRNA transfection or ADMA treatment than controls. Addition of an NO donor restored cleaved caspase‐3 and ‐8 protein levels after DDAH1 siRNA transfection in hfPMVEC to essentially the levels seen in scramble control. Addition of a putative caspase‐3 inhibitor to DDAH1 siRNA transfected and NO‐donor treated cells led to greater numbers of viable cells and far less angiogenesis than in any other group studied. We conclude that in hfPMVEC, DDAH1 is central to the regulation of NO‐mediated caspase‐3 activation and the resultant apoptosis and angiogenesis. Our findings suggest that DDAH1 may be a potential therapeutic target in pulmonary hypertensive disorders.
Collapse
Affiliation(s)
- Jennifer K Trittmann
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Hanadi Almazroue
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Yi Jin
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Leif D Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
6
|
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
|
7
|
Pool CM, Jin Y, Chen B, Liu Y, Nelin LD. Hypoxic-induction of arginase II requires EGF-mediated EGFR activation in human pulmonary microvascular endothelial cells. Physiol Rep 2018; 6:e13693. [PMID: 29845760 PMCID: PMC5974731 DOI: 10.14814/phy2.13693] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 03/31/2018] [Accepted: 04/03/2018] [Indexed: 12/22/2022] Open
Abstract
We have previously shown that hypoxia-induced proliferation of human pulmonary microvascular endothelial cells (hPMVEC) depends on arginase II, and that epidermal growth factor receptor (EGFR) is necessary for hypoxic-induction of arginase II. However, it remains unclear how hypoxia activates EGFR-mediated signaling in hPMVEC. We hypothesized that hypoxia results in epidermal growth factor (EGF) production and that EGF binds to EGFR to activate the signaling cascade leading to arginase II induction and proliferation in hPMVEC. We found that hypoxia significantly increased the mRNA levels of EGF, EGFR, and arginase in hPMVEC. Hypoxia significantly increased pEGFR(Tyr845) protein levels and an EGF neutralizing antibody prevented the hypoxic induction of pEGFR. Inhibiting EGFR activation prevented hypoxia-induced arginase II mRNA and protein induction. Treatment of hPMVEC with exogenous EGF resulted in greater levels of arginase II protein both in normoxia and hypoxia. An EGF neutralizing antibody diminished hypoxic induction of arginase II and resulted in fewer viable cells in hPMVEC. Similarly, siRNA against EGF prevented hypoxic induction of arginase II and resulted in fewer viable cells. Finally, conditioned media from hypoxic hPMVEC induced proliferation in human pulmonary artery smooth muscle cells (hPASMC), however, conditioned media from a group of hypoxic hPMVEC in which EGF were knocked down did not promote hPASMC proliferation. These findings demonstrate that hypoxia-induced arginase II expression and cellular proliferation depend on autocrine EGF production leading to EGFR activation in hPMVEC. We speculate that EGF-EGFR signaling may be a novel therapeutic target for pulmonary hypertensive disorders associated with hypoxia.
Collapse
Affiliation(s)
- Caitlyn M. Pool
- Pulmonary Hypertension GroupCenter for Perinatal ResearchResearch Institute at Nationwide Children's HospitalColumbusOhio
- Department of PediatricsThe Ohio State UniversityColumbusOhio
| | - Yi Jin
- Pulmonary Hypertension GroupCenter for Perinatal ResearchResearch Institute at Nationwide Children's HospitalColumbusOhio
- Department of PediatricsThe Ohio State UniversityColumbusOhio
| | - Bernadette Chen
- Pulmonary Hypertension GroupCenter for Perinatal ResearchResearch Institute at Nationwide Children's HospitalColumbusOhio
- Department of PediatricsThe Ohio State UniversityColumbusOhio
| | - Yusen Liu
- Pulmonary Hypertension GroupCenter for Perinatal ResearchResearch Institute at Nationwide Children's HospitalColumbusOhio
- Department of PediatricsThe Ohio State UniversityColumbusOhio
| | - Leif D. Nelin
- Pulmonary Hypertension GroupCenter for Perinatal ResearchResearch Institute at Nationwide Children's HospitalColumbusOhio
- Department of PediatricsThe Ohio State UniversityColumbusOhio
| |
Collapse
|
8
|
Trittmann JK, Velten M, Heyob KM, Almazroue H, Jin Y, Nelin LD, Rogers LK. Arginase and α-smooth muscle actin induction after hyperoxic exposure in a mouse model of bronchopulmonary dysplasia. Clin Exp Pharmacol Physiol 2018; 45:556-562. [PMID: 29266319 DOI: 10.1111/1440-1681.12909] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/30/2017] [Accepted: 12/12/2017] [Indexed: 12/26/2022]
Abstract
The L-arginine/NO pathway is an important regulator of pulmonary hypertension, the leading cause of mortality in patients with the chronic lung disease of prematurity, bronchopulmonary dysplasia. L-arginine can be metabolized by NO synthase (NOS) to form L-citrulline and NO, a potent vasodilator. Alternatively, L-arginine can be metabolized by arginase to form urea and L-ornithine, a precursor to collagen and proline formation important in vascular remodelling. In the current study, we hypothesized that C3H/HeN mice exposed to prolonged hyperoxia would have increased arginase expression and pulmonary vascular wall cell proliferation. C3H/HeN mice were exposed to 14 days of 85% O2 or room air and lung homogenates analyzed by western blot for protein levels of arginase I, arginase II, endothelial NOS (eNOS), ornithine decarboxylase (ODC), ornithine aminotransferase (OAT), and α-smooth muscle actin (α-SMA). Hyperoxia did not change arginase I or eNOS protein levels. However, arginase II protein levels were 15-fold greater after hyperoxia exposure than in lungs exposed to room air. Greater protein levels of ODC and OAT were found in lungs following hyperoxic exposure than in room air animals. α-SMA protein levels were found to be 7-fold greater in the hyperoxia exposed lungs than in room air lungs. In the hyperoxia exposed lungs there was evidence of greater pulmonary vascular wall cell proliferation by α-SMA immunohistochemistry than in room air lungs. Taken together, these data are consistent with a more proliferative vascular phenotype, and may explain the propensity of patients with bronchopulmonary dysplasia to develop pulmonary hypertension.
Collapse
Affiliation(s)
- Jennifer K Trittmann
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Markus Velten
- Department of Anesthesiology and Intensive Care Medicine, Rheinische Friedrich-Wilhelms University, University Medical Center, Bonn, Germany
| | - Kathryn M Heyob
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Hanadi Almazroue
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Yi Jin
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Leif D Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | - Lynette K Rogers
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
9
|
White HA, Jin Y, Chicoine LG, Chen B, Liu Y, Nelin LD. Hypoxic proliferation requires EGFR-mediated ERK activation in human pulmonary microvascular endothelial cells. Am J Physiol Lung Cell Mol Physiol 2017; 312:L649-L656. [PMID: 28188223 DOI: 10.1152/ajplung.00267.2016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 02/07/2017] [Accepted: 02/07/2017] [Indexed: 02/06/2023] Open
Abstract
We have previously shown that hypoxic proliferation of human pulmonary microvascular endothelial cells (hPMVECs) depends on epidermal growth factor receptor (EGFR) activation. To determine downstream signaling leading to proliferation, we tested the hypothesis that hypoxia-induced proliferation in hPMVECs would require EGFR-mediated activation of extracellular signal-regulated kinase (ERK) leading to arginase II induction. To test this hypothesis, hPMVECs were incubated in either normoxia (21% O2, 5% CO2) or hypoxia (1% O2, 5% CO2) and Western blotting was performed for EGFR, arginase II, phosphorylated-ERK (pERK), and total ERK (ERK). Hypoxia led to greater EGFR, pERK, and arginase II protein levels than did normoxia in hPMVECs. To examine the role of EGFR in these hypoxia-induced changes, hPMVECs were transfected with siRNA against EGFR or a scrambled siRNA and placed in hypoxia. Inhibition of EGFR using siRNA attenuated hypoxia-induced pERK and arginase II expression as well as the hypoxia-induced increase in viable cell numbers. hPMVECs were then treated with vehicle, an EGFR inhibitor (AG1478), or an ERK pathway inhibitor (U0126) and placed in hypoxia. Pharmacologic inhibition of EGFR significantly attenuated the hypoxia-induced increase in pERK level. Both AG1478 and U0126 also significantly attenuated the hypoxia-induced increase in viable hPMVECs numbers. hPMVECs were transfected with an adenoviral vector containing arginase II (AdArg2) and overexpression of arginase II rescued the U0126-mediated decrease in viable cell numbers in hypoxic hPMVECs. Our findings suggest that hypoxic activation of EGFR results in phosphorylation of ERK, which is required for hypoxic induction of arginase II and cellular proliferation.
Collapse
Affiliation(s)
- Hilary A White
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Yi Jin
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Louis G Chicoine
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Yusen Liu
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and.,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Leif D Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, Columbus, Ohio; and .,Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
10
|
Ahmad AS, Shah ZA, Doré S. Protective Role of Arginase II in Cerebral Ischemia and Excitotoxicity. ACTA ACUST UNITED AC 2016; 7. [PMID: 27308186 DOI: 10.21767/2171-6625.100088] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Arginase (Arg), one of the enzymes involved in the urea cycle, provides an essential route for the disposal of excess nitrogen resulting from amino acid and nucleotide metabolism. Two reported subtypes of Arg (ArgI and II) compete with nitric oxide synthase (NOS) to use L-arginine as a substrate, and subsequently regulate NOS activity. It has been reported that Arg has significant effects on circulation that suggest the potential role of this enzyme in regulating vascular function. However, the role of Arg following brain damage has not been elucidated. In this study, we hypothesize that the deletion of ArgII will lead to aggravated brain injury following cerebral ischemia and excitotoxicity. METHODS AND FINDINGS To test our hypothesis, male C57BL/6 wildtype (WT) and ArgII-/- mice were subjected to permanent distal middle cerebral artery occlusion and survived for 7 d. Cerebral blood flow (CBF) data revealed a statistically non-significant decrease in CBF in ArgII-/- mice. However, ArgII-/- mice had significantly higher neurologic deficit scores and brain infarctions. The hypothesis was further tested in a more specific N-methyl-D-aspartate (NMDA)-induced acute excitotoxic model. WT and ArgII-/- mice were given a single intrastriatal injection of 15 nmol NMDA. Forty-eight hours later, the excitotoxic brain damage was significantly worse in ArgII-/- mice. The data from both models confirm the neuroprotective effect of ArgII. CONCLUSION Targeting ArgII could be considered an integrative part of a multi-modal approach to fight acute brain damage excitotoxicity, ischemic brain injury, and other forms of brain trauma.
Collapse
Affiliation(s)
- Abdullah Shafique Ahmad
- Department of Anesthesiology, University of Florida, Gainesville, 32610, FL, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, 32610, FL, USA
| | - Zahoor Ahmad Shah
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo 43614, OH, USA
| | - Sylvain Doré
- Department of Anesthesiology, University of Florida, Gainesville, 32610, FL, USA; Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, 32610, FL, USA; Departments of Neurology, Psychiatry, Psychology, Pharmaceutics, and Neuroscience, University of Florida, Gainesville, 32610 FL, USA
| |
Collapse
|
11
|
Chu Y, XiangLi X, Niu H, Wang H, Jia P, Gong W, Wu D, Qin W, Xing C. Arginase inhibitor attenuates pulmonary artery hypertension induced by hypoxia. Mol Cell Biochem 2016; 412:91-9. [PMID: 26608181 DOI: 10.1007/s11010-015-2611-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/14/2015] [Indexed: 02/06/2023]
Abstract
Hypoxia-induced pulmonary arterial hypertension (HPAH) is a refractory disease characterized by increased proliferation of pulmonary vascular smooth cells and progressive pulmonary vascular remodeling. The level of nitric oxide (NO), a potential therapeutic vasodilator, is low in PAH patients. L-arginine can be converted to either beneficial NO by nitric oxide synthases or to harmful urea by arginase. In the present study, we aimed to investigate whether an arginase inhibitor, S-(2-boronoethyl)-L-cysteine ameliorates HPAH in vivo and vitro. In a HPAH mouse model, we assessed right ventricle systolic pressure (RVSP) by an invasive method, and found that RSVP was elevated under hypoxia, but was attenuated upon arginase inhibition. Human pulmonary artery smooth muscle cells (HPASMCs) were cultured under hypoxic conditions, and their proliferative capacity was determined by cell counting and flow cytometry. The levels of cyclin D1, p27, p-Akt, and p-ERK were detected by RT-PCR or Western blot analysis. Compared to hypoxia group, arginase inhibitor inhibited HPASMCs proliferation and reduced the levels of cyclin D1, p-Akt, p-ERK, while increasing p27 level. Moreover, in mouse models, compared to control group, hypoxia increased cyclin D1 expression but reduced p27 expression, while arginase inhibitor reversed the effects of hypoxia. Taken together, these results suggest that arginase plays an important role in increased proliferation of HPASMCs induced by hypoxia and it is a potential therapeutic target for the treatment of pulmonary hypertensive disorders.
Collapse
Affiliation(s)
- YanBiao Chu
- Department of Respiration, Jinan Central Hospital Affiliated to Shandong University, 105 JieFang Rd, Ji'nan, 250013, Shandong, China
| | - XiaoYing XiangLi
- Department of Surgery, Qilu Hospital, Shandong University, Ji'nan, 250012, Shandong, China
| | - Hu Niu
- Department of General Surgery, The Fourth People's Hospital of Ji'nan, The Second Affiliated Hospital of Tai Shan Medical College, Ji'nan, 250031, China
| | - HongChao Wang
- Department of Respiration, Jinan Central Hospital Affiliated to Shandong University, 105 JieFang Rd, Ji'nan, 250013, Shandong, China
| | - PingDong Jia
- Department of Respiration, Jinan Central Hospital Affiliated to Shandong University, 105 JieFang Rd, Ji'nan, 250013, Shandong, China
| | - WenBin Gong
- Department of Respiration, Jinan Central Hospital Affiliated to Shandong University, 105 JieFang Rd, Ji'nan, 250013, Shandong, China
| | - DaWei Wu
- Department of Critical Care Medicine, Qilu Hospital, Shandong University, Ji'nan, 250012, Shandong, China
| | - WeiDong Qin
- Department of Critical Care Medicine, Qilu Hospital, Shandong University, Ji'nan, 250012, Shandong, China
| | - ChunYan Xing
- Department of Respiration, Jinan Central Hospital Affiliated to Shandong University, 105 JieFang Rd, Ji'nan, 250013, Shandong, China.
| |
Collapse
|
12
|
Chen B, Strauch K, Jin Y, Cui H, Nelin LD, Chicoine LG. Asymmetric dimethylarginine does not inhibit arginase activity and is pro-proliferative in pulmonary endothelial cells. Clin Exp Pharmacol Physiol 2015; 41:469-74. [PMID: 24799070 DOI: 10.1111/1440-1681.12252] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 04/18/2014] [Accepted: 04/24/2014] [Indexed: 11/27/2022]
Abstract
Asymmetric dimethylarginine (ADMA) is an endogenously produced nitric oxide synthase (NOS) inhibitor. L-Arginine can be metabolised by NOS and arginase, and arginase is the first step in polyamine production necessary for cellular proliferation. We tested the hypothesis that ADMA would inhibit NOS but not arginase activity and that this pattern of inhibition would result in greater L-arginine bioavailability to arginase, thereby increasing viable cell number. Bovine arginase was used in in vitro activity assays with various concentrations of substrate (L-arginine, ADMA, N(G) -monomethyl-L-arginine (L-NMMA) and N(G) -nitro-L-arginine methyl ester (L-NAME)). Only L-arginine resulted in measurable urea production (Km = 6.9 ± 0.8 mmol/L; Vmax = 6.6 ± 0.3 μmol/mg protein per min). We then incubated bovine arginase with increasing concentrations of ADMA, L-NMMA and L-NAME in the presence of 1 mmol/L l-arginine and found no effect of any of the tested compounds on arginase activity. Using bovine pulmonary arterial endothelial cells (bPAEC) we determined the effects of ADMA on nitric oxide (NO) and urea production and found significantly lower NO production and greater urea production (P < 0.003) with ADMA, without changes in arginase protein levels. In addition, ADMA treatment resulted in an approximately 30% greater number of viable cells after 48 h than in control bPAEC. These results demonstrate that ADMA is neither a substrate nor an inhibitor of arginase activity and that in bPAEC ADMA inhibits NO production and enhances urea production, leading to more viable cells. These results may have pathophysiological implications in disorders associated with higher ADMA levels, such as pulmonary hypertension.
Collapse
Affiliation(s)
- Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | | | | | | | | | | |
Collapse
|
13
|
Jin Y, Pang T, Nelin LD, Wang W, Wang Y, Yan J, Zhao C. MKP-1 is a target of miR-210 and mediate the negative regulation of miR-210 inhibitor on hypoxic hPASMC proliferation. Cell Biol Int 2014; 39:113-20. [PMID: 25044272 DOI: 10.1002/cbin.10339] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 06/25/2014] [Indexed: 11/06/2022]
Abstract
Chonic hypoxia, smooth muscle cell (SMC) proliferation and vascular remodeling are hallmark features of pathogenic pulmonary artery hypertension. MicroRNAs (miRNAs), endogenously expressed small noncoding RNAs, regulate gene expression at the post-transcriptional level. MiR-210 is considered a "master miRNA" in the control of diverse functions in hypoxic cells and tissues and has a cytoprotective function in pulmonary artery SMCs during hypoxic stress. MiR-210 is also upregulated in lung tissue of chonically hypoxic mice suffering from pulmonary hypertension. Jin et al. () showed that mice deficient in mitogen-activated protein kinase phosphatase 1 (MKP-1) had severe hypoxia-induced pulmonary hypertension, so MKP-1 may be important in the progression of hypoxic pulmonary artery hypertension. We investigated the possible interactions between miR-210 and MKP-1 and the effect on cell proliferation in hypoxic human pulmonary artery SMCs (hPASMCs). miR-210 was significantly increased in cultured hPASMCs exposed to 1% O2 hypoxia for 48 h, as was MKP-1 mRNA and protein expression. Furthermore, inhibiting miR-210 expression increased MKP-1 mRNA and protein expression in hPASMCs and decreased cell proliferation under hypoxia. Conversely, overexpressing miR-210 prevented hypoxia-induced MKP-1 expression with no effect on cell proliferation. siRNA knockdown of MKP-1 abolished the miR-210-inhibition prevention of cell proliferation under hypoxia. MKP-1 is a target of miR-210 and could mediate the negative regulation of miR-210 inhibition on hypoxic hPASMCs.
Collapse
Affiliation(s)
- Youpeng Jin
- Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, Shandong, China
| | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
PURPOSE OF THE REVIEW The epidermal growth factor receptor (EGFR) is a receptor tyrosine kinase with a wide implication in tumor biology, wound healing and development. Besides acting as a growth factor receptor activated by ligands such as EGF, the EGFR can also be transactivated and thereby mediate cross-talk with different signaling pathways. The aim of this review is to illustrate the Janus-faced function of the EGFR in the vasculature with its relevance for vascular biology and disease. RECENT FINDINGS Over recent years, the number of identified signaling partners of the EGFR has steadily increased, as have the biological processes in which the EGFR is thought to be involved. Recently, new models have allowed investigation of EGFR effects in vivo, shedding some light on the overall function of the EGFR in the vasculature. At the same time, EGFR inhibitors and antibodies have become increasingly established in cancer therapy, providing potential therapeutic tools for decreasing EGFR signaling. SUMMARY The EGFR is a versatile signaling pathway integrator associated with vascular homeostasis and disease. In addition to modulating basal vascular tone and tissue homeostasis, the EGFR also seems to be involved in proinflammatory, proliferative, migratory and remodeling processes, with enhanced deposition of extracellular matrix components, thereby promoting vascular diseases such as hypertension or atherosclerosis.
Collapse
|
15
|
Jin Y, Jin Y, Chen B, Tipple TE, Nelin LD. Arginase II is a target of miR-17-5p and regulates miR-17-5p expression in human pulmonary artery smooth muscle cells. Am J Physiol Lung Cell Mol Physiol 2014; 307:L197-204. [PMID: 24879052 DOI: 10.1152/ajplung.00266.2013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Vascular remodeling and smooth muscle cell proliferation are hallmark pathogenic features of pulmonary artery hypertension. MicroRNAs, endogenously expressed small noncoding RNAs, regulate gene expression at the posttranscriptional level. It has previously been shown that miR-17 overexpression in cultured human pulmonary artery smooth muscle cell (hPASMC) resulted in increased viable cell number. Previously, we have found that arginase II promotes hypoxia-induced proliferation in hPASMC. Therefore, we hypothesized that miR-17 would be upregulated by hypoxia in hPASMC and would result in greater arginase II expression. We found that levels of miR-17-5p and arginase II were significantly greater in cultured hPASMC exposed to 1% O2 for 48 h than in hPASMC exposed to 21% O2 for 48 h. Furthermore, inhibiting miR-17-5p expression decreased hypoxia-induced arginase II protein levels in hPASMC. Conversely, overexpressing miR-17-5p resulted in greater arginase II protein levels. Somewhat surprisingly, arginase II inhibition was associated with lower miR-17-5p expression in both normoxic and hypoxic hPASMC, whereas overexpressing arginase II resulted in greater miR-17-5p expression in hPASMC. These findings suggest that hypoxia-induced arginase II expression is not only regulated by miR-17-5p but also that there is a feedback loop between arginase II and miR-17-5p in hPASMC. We also found that the arginase II-mediated regulation of miR-17-5p was independent of either p53 or c-myc. We also found that l-arginine, the substrate for arginase II, and l-ornithine, the amino acid product of arginase II, were not involved in the regulation of miR-17-5p expression.
Collapse
Affiliation(s)
- Youpeng Jin
- Pediatric Intensive Care Unit, Provincial Hospital affiliated to Shandong University, Jinan, Shandong, China; and Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Yi Jin
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Trent E Tipple
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University, Columbus, Ohio
| | - Leif D Nelin
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital and Department of Pediatrics, The Ohio State University, Columbus, Ohio
| |
Collapse
|
16
|
Lou Y, Zhang G, Geng M, Zhang W, Cui J, Liu S. TIPE2 negatively regulates inflammation by switching arginine metabolism from nitric oxide synthase to arginase. PLoS One 2014; 9:e96508. [PMID: 24806446 PMCID: PMC4013027 DOI: 10.1371/journal.pone.0096508] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 04/09/2014] [Indexed: 12/28/2022] Open
Abstract
TIPE2, the tumor necrosis factor (TNF)-alpha-induced protein 8-like 2 (TNFAIP8L2), plays an essential role in maintaining immune homeostasis. It is highly expressed in macrophages and negatively regulates inflammation through inhibiting Toll-like receptor signaling. In this paper, we utilized RAW264.7 cells stably transfected with a TIPE2 expression plasmid, as well as TIPE2-deficient macrophages to study the roles of TIPE2 in LPS-induced nitric oxide (NO) and urea production. The results showed that TIPE2-deficiency significantly upregulated the levels of iNOS expression and NO production in LPS-stimulated macrophages, but decreased mRNA levels of arginase I and urea production. However, TIPE2 overexpression in macrophages was capable of downregulating protein levels of LPS-induced iNOS and NO, but generated greater levels of arginase I and urea production. Furthermore, TIPE2−/− mice had higher iNOS protein levels in lung and liver and higher plasma NO concentrations, but lower levels of liver arginase I compared to LPS-treated WT controls. Interestingly, significant increases in IκB degradation and phosphorylation of JNK, p38, and IκB were observed in TIPE2-deficient macrophages following LPS challenge. These results strongly suggest that TIPE2 plays an important role in shifting L-arginase metabolism from production of NO to urea, during host inflammatory response.
Collapse
Affiliation(s)
- Yunwei Lou
- Department of Immunology, Shandong University School of Medicine, Ji'nan, P.R. China
| | - Guizhong Zhang
- Department of Immunology, Shandong University School of Medicine, Ji'nan, P.R. China
| | - Minghong Geng
- Department of Immunology, Shandong University School of Medicine, Ji'nan, P.R. China
| | - Wenqian Zhang
- Department of Immunology, Shandong University School of Medicine, Ji'nan, P.R. China
| | - Jian Cui
- Department of Immunology, Shandong University School of Medicine, Ji'nan, P.R. China
| | - Suxia Liu
- Department of Immunology, Shandong University School of Medicine, Ji'nan, P.R. China
- * E-mail:
| |
Collapse
|
17
|
Bulgaroni V, Lombardo P, Rivero-Osimani V, Vera B, Dulgerian L, Cerbán F, Rivero V, Magnarelli G, Guiñazú N. Environmental pesticide exposure modulates cytokines, arginase and ornithine decarboxylase expression in human placenta. Reprod Toxicol 2013; 39:23-32. [DOI: 10.1016/j.reprotox.2013.03.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 03/04/2013] [Accepted: 03/21/2013] [Indexed: 11/24/2022]
|
18
|
Abstract
Arginase metabolizes the semi-essential amino acid l-arginine to l-ornithine and urea. There are two distinct isoforms of arginase, arginase I and II, which are encoded by separate genes and display differences in tissue distribution, subcellular localization, and molecular regulation. Blood vessels express both arginase I and II but their distribution appears to be cell-, vessel-, and species-specific. Both isoforms of arginase are induced by numerous pathologic stimuli and contribute to vascular cell dysfunction and vessel wall remodeling in several diseases. Clinical and experimental studies have documented increases in the expression and/or activity of arginase I or II in blood vessels following arterial injury and in pulmonary and arterial hypertension, aging, and atherosclerosis. Significantly, pharmacological inhibition or genetic ablation of arginase in animals ameliorates abnormalities in vascular cells and normalizes blood vessel architecture and function in all of these pathological states. The detrimental effect of arginase in vascular remodeling is attributable to its ability to stimulate vascular smooth muscle cell and endothelial cell proliferation, and collagen deposition by promoting the synthesis of polyamines and l-proline, respectively. In addition, arginase adversely impacts arterial remodeling by directing macrophages toward an inflammatory phenotype. Moreover, the proliferative, fibrotic, and inflammatory actions of arginase in the vasculature are further amplified by its capacity to inhibit nitric oxide (NO) synthesis by competing with NO synthase for substrate, l-arginine. Pharmacologic or molecular approaches targeting specific isoforms of arginase represent a promising strategy in treating obstructive fibroproliferative vascular disease.
Collapse
Affiliation(s)
- William Durante
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia Columbia, MO, USA
| |
Collapse
|
19
|
Cui H, Chen B, Chicoine LG, Nelin LD. Overexpression of cationic amino acid transporter-1 increases nitric oxide production in hypoxic human pulmonary microvascular endothelial cells. Clin Exp Pharmacol Physiol 2012; 38:796-803. [PMID: 21923750 DOI: 10.1111/j.1440-1681.2011.05609.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
1. The endogenous production of and/or the bioavailability of nitric oxide (NO) is decreased in pulmonary hypertensive diseases. L-arginine (L-arg) is the substrate for NO synthase (NOS). L-arg is transported into cells via the cationic amino acid transporters (CAT), of which there are two isoforms in endothelial cells, CAT-1 and CAT-2. 2. To test the hypothesis that hypoxia will decrease CAT expression and L-arg uptake resulting in decreased NO production in human pulmonary microvascular endothelial cells (hPMVEC), cells were incubated in either normoxia (21% O(2), 5% CO(2), balance N(2)) or hypoxia (1% O(2), 5% CO(2), balance N(2)). 3. The hPMVEC incubated in hypoxia had 80% less NO production than cells incubated in normoxia (P < 0.01). The hPMVEC incubated in hypoxia had significantly lower CAT-2 mRNA levels than normoxic hPMVEC (P < 0.005), and the transport of L-arg was 40% lower in hypoxic than in normoxic hPMVEC (P < 0.01). In hypoxic cells, overexpression of CAT-1 resulted in significantly greater L-arg transport and NO production (P < 0.05). 4. These results demonstrate that in hPMVEC, hypoxia decreased CAT-2 expression, L-arg uptake and NO production. Furthermore, the hypoxia-induced decrease in NO production in hPMVEC can be attenuated by overexpressing CAT in these cells. We speculate that the CAT may represent a novel therapeutic target for treating pulmonary hypertensive disorders.
Collapse
Affiliation(s)
- Hongmei Cui
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University, Columbus, OH, USA
| | | | | | | |
Collapse
|
20
|
Erythropoietin alleviates post-ischemic injury of rat hearts by attenuating nitrosative stress. Life Sci 2012; 90:776-84. [PMID: 22521289 DOI: 10.1016/j.lfs.2012.04.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 03/14/2012] [Accepted: 04/03/2012] [Indexed: 11/22/2022]
Abstract
AIMS Nitrosative stress caused by ischemia contributes to poor functional recovery in hearts. A previous study showed that recombinant human erythropoietin (EPO) activates the Janus-tyrosine kinase 2/extracellular signal-regulated kinase (Jak2/ERK) pathway to protect myocardium against ischemia/reperfusion (IR) injury. However, it is not clear how pro-survival signals triggered by EPO affect the nitric oxide (NO) system in post-ischemic myocardial tissue. MAIN METHODS Isolated rat hearts were subjected to IR injury and changes in protein expression in the myocardium were evaluated by immunostaining. KEY FINDINGS Compared with untreated hearts, EPO-treated IR hearts showed significant improvements in contractility and reduced myocardial injury and infarction; this was associated with attenuated caspase-3 activation. Excess formation of NO metabolites and nitrotyrosine, which cause nitrosative stress, was markedly suppressed by EPO. The mechanism underlying EPO-mediated alleviation of nitrosative stress was related to an increase in arginase II expression and to the suppression of heat shock protein 90 (HSP90)-dependent upregulation of endothelial and inducible NO synthase (NOS). Myocardial EPO content was restored after EPO treatment, which in turn recruited signal transducer and activator of transcription (STAT) 3 protein and induced ERK signaling downstream of Jak2, which increased arginase II levels and suppressed HSP90 expression, respectively. Inhibition of STAT3 and ERK specifically reversed the effects of EPO on arginase II and HSP90 expression. SIGNIFICANCE These results indicate that EPO triggers the Jak2-STAT3/ERK pathway to restore the balance between arginase and NOS and, thus, reduces nitrosative stress. This may form the basis of myocardial protection following IR.
Collapse
|
21
|
Chen B, Calvert AE, Meng X, Nelin LD. Pharmacologic agents elevating cAMP prevent arginase II expression and proliferation of pulmonary artery smooth muscle cells. Am J Respir Cell Mol Biol 2012; 47:218-26. [PMID: 22447968 DOI: 10.1165/rcmb.2011-0015oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Arginase II has been shown to be involved in the hypoxia-induced proliferation of human pulmonary artery smooth muscle cells (hPASMCs). The signal transduction pathways responsible for the induction of arginase II are poorly understood. Cyclic AMP is involved in many intracellular processes, and cAMP levels are regulated by a balance between production via adenylate cyclases and degradation via phosphodiesterases. The purpose of this study was to determine the effects of cAMP on hypoxia-induced arginase expression, activity, and proliferation in hPASMCs. We found that the cAMP analog 8-Bromo-cAMP (8-Br-cAMP), the adenylate cyclase activator forskolin, and the phosphodiesterase 3 inhibitor cilostamide prevented the hypoxic induction of arginase II mRNA and protein expression in hPASMCs. The inhibition of arginase II protein was found to be mediated by exchange protein directly activated by cAMP. Arginase activity was decreased by 8-Br-cAMP, as evidenced by significantly lower V(max) for arginase in normoxia and hypoxia. The hypoxia-induced hPASMC proliferation was completely prevented by the addition of 8-Br-cAMP, forskolin, or cilostamide. These data are the first to describe the inhibitory effect of cAMP on arginase activity, expression, and resultant proliferation of hypoxic hPASMCs.
Collapse
Affiliation(s)
- Bernadette Chen
- Pulmonary Hypertension Group, Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA.
| | | | | | | |
Collapse
|
22
|
Perez G, Olivares IM, Rodriguez MG, Ceballos GM, Garcia Sanchez JR. Arginase Activity in Patients with Breast Cancer: An Analysis of Plasma, Tumors, and Its Relationship with the Presence of the Estrogen Receptor. ACTA ACUST UNITED AC 2012; 35:570-4. [DOI: 10.1159/000343005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
23
|
Hyseni X, Soukup JM, Huang YCT. Pollutant particles induce arginase II in human bronchial epithelial cells. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2012; 75:624-636. [PMID: 22712848 DOI: 10.1080/15287394.2012.688479] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Exposure to particulate matter (PM) is associated with adverse pulmonary effects, including induction and exacerbation of asthma. Recently arginase was shown to play an important role in the pathogenesis of asthma. In this study, it was postulated that PM exposure might induce arginase. Human bronchial epithelial cells (HBEC) obtained from normal individuals by endobronchial brushings cultured on an air-liquid interface were incubated with fine Chapel Hill particles (PM₂.₅, 100 μg/ml) for up to 72 h. Arginase activity, protein expression, and mRNA of arginase I and arginase II were measured. PM₂.₅ increased arginase activity in a time-dependent manner. The rise was primarily due to upregulation of arginase II. PD153035 (10 μM), an epidermal growth factor (EGF) receptor antagonist, attenuated the PM₂.₅-induced elevation in arginase activity and arginase II expression. Treatment of HBEC with human EGF increased arginase activity and arginase II expression. Pretreatment with catalase (200 U/ml), superoxide dismutase (100 U/ml), or apocynin (5 μg/ml), an NAD(P)H oxidase inhibitor, did not markedly affect arginase II expression. Treatment of HBEC with arginase II siRNA inhibited the expression of arginase II by 60% and increased IL-8 release induced by PM₂.₅. These results indicate that PM exposure upregulates arginase II activity and expression in human bronchial epithelial cells, in part via EGF-dependent mechanisms independent of oxidative stress. The elevated arginase II activity and expression may be a mechanism underlying adverse effects induced by PM exposure in asthma patients.
Collapse
Affiliation(s)
- Xhevahire Hyseni
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina, USA
| | | | | |
Collapse
|
24
|
Prati C, Berthelot A, Wendling D, Demougeot C. Endothelial dysfunction in rat adjuvant-induced arthritis: Up-regulation of the vascular arginase pathway. ACTA ACUST UNITED AC 2011; 63:2309-17. [DOI: 10.1002/art.30391] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
25
|
Zuckerbraun BS, George P, Gladwin MT. Nitrite in pulmonary arterial hypertension: therapeutic avenues in the setting of dysregulated arginine/nitric oxide synthase signalling. Cardiovasc Res 2010; 89:542-52. [PMID: 21177703 DOI: 10.1093/cvr/cvq370] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is an insidious disease of the small pulmonary arteries that is progressive in nature and results in right heart strain/hypertrophy and eventually failure. The aetiologies may vary but several common pathophysiological changes result in this phenotype, including vasoconstriction, thrombosis, and vascular proliferation. Data suggest that nitric oxide (NO) signalling is vasoprotective in the setting of PAH. The classic arginine-NO synthase (NOS)-NO signalling pathway may represent an adaptive response that is eventually dysregulated during disease progression. Dysregulation occurs secondary to NOS enzyme down-regulation, enzymatic uncoupling, and arginine catabolism by vascular and red cell arginases and by direct NO inactivation via catabolic reactions with superoxide or cell-free plasma haemoglobin (in the case of haemolytic disease). The anion nitrite, which has recently been recognized as a source of NO that circumvents the arginine-NOS pathway, may serve as an additional adaptive signalling pathway that is now appreciated to have a vasoregulatory role in the pulmonary and systemic vasculature. Inhaled nebulized sodium nitrite is a relatively potent pulmonary vasodilator in the setting of hypoxia and is also anti-proliferative in multiple experimental models of pulmonary hypertension. Multiple nitrite reductases have been shown to be relevant in the conversion of nitrite to metabolically active NO, including deoxy-haemoglobin and myoglobin in the circulation and heart, respectively, and xanthine oxidoreductase in the lung parenchyma.
Collapse
Affiliation(s)
- Brian S Zuckerbraun
- Department of Surgery, University of Pittsburgh, NW 607 MUH, 3459 Fifth Avenue, Pittsburgh, PA 15213, USA.
| | | | | |
Collapse
|
26
|
Inhaled nitric oxide prevents 3-nitrotyrosine formation in the lungs of neonatal mice exposed to >95% oxygen. Lung 2010; 188:217-27. [PMID: 20237791 DOI: 10.1007/s00408-010-9235-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Accepted: 03/02/2010] [Indexed: 10/19/2022]
Abstract
Inhaled nitric oxide is being evaluated as a preventative therapy for patients at risk for bronchopulmonary dysplasia (BPD). Nitric oxide (NO), in the presence of superoxide, forms peroxynitrite, which reacts with tyrosine residues on proteins to form 3-nitrotyrosine (3-NT). However, NO can also act as an antioxidant and was recently found to improve the oxidative balance in preterm infants. Thus, we tested the hypothesis that the addition of a therapeutically relevant concentration (10 ppm) of NO to a hyperoxic exposure would lead to decreased 3-NT formation in the lung. FVB mouse pups were exposed to either room air (21% O(2)) or >95% O(2) with or without 10 ppm NO within 24 h of birth. In the first set of studies, body weights and survival were monitored for 7 days, and exposure to >95% O(2) resulted in impaired weight gain and near 100% mortality by 7 days. However, the mortality occurred earlier in pups exposed to >95% O(2) + NO than in pups exposed to >95% O(2) alone. In a second set of studies, lungs were harvested at 72 h. Immunohistochemistry of the lungs at 72 h revealed that the addition of NO decreased alveolar, bronchial, and vascular 3-NT staining in pups exposed to both room air and hyperoxia. The lung nitrite levels were higher in animals exposed to >95% oxygen + NO than in animals exposed to >95% oxygen alone. The protein levels of myeloperoxidase, monocyte chemotactic protein-1, and intracellular adhesion molecule-1 were assessed after 72 h of exposure and found to be greatest in the lungs of pups exposed to >95% O(2). This hyperoxia-induced protein expression was significantly attenuated by the addition of 10 ppm NO. We propose that in the presence of >95% O(2), peroxynitrite formation results in protein nitration; however, adding excess NO to the >95% O(2) exposure prevents 3-NT formation by NO reacting with peroxynitrite to produce nitrite and NO(2). We speculate that the decreased protein nitration observed with the addition of NO may be a potential mechanism limiting hyperoxic lung injury.
Collapse
|
27
|
Jin Y, Calvert TJ, Chen B, Chicoine LG, Joshi M, Bauer JA, Liu Y, Nelin LD. Mice deficient in Mkp-1 develop more severe pulmonary hypertension and greater lung protein levels of arginase in response to chronic hypoxia. Am J Physiol Heart Circ Physiol 2010; 298:H1518-28. [PMID: 20173047 DOI: 10.1152/ajpheart.00813.2009] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The mitogen-activated protein (MAP) kinases are involved in cellular responses to many stimuli, including hypoxia. MAP kinase signaling is regulated by a family of phosphatases that include MAP kinase phosphatase-1 (MKP-1). We hypothesized that mice lacking the Mkp-1 gene would have exaggerated chronic hypoxia-induced pulmonary hypertension. Wild-type (WT) and Mkp-1(-/-) mice were exposed to either 4 wk of normoxia or hypobaric hypoxia. Following chronic hypoxia, both genotypes demonstrated elevated right ventricular pressures, right ventricular hypertrophy as demonstrated by the ratio of the right ventricle to the left ventricle plus septum weights [RV(LV + S)], and greater vascular remodeling. However, the right ventricular systolic pressures, the RV/(LV + S), and the medial wall thickness of 100- to 300-microm vessels was significantly greater in the Mkp-1(-/-) mice than in the WT mice following 4 wk of hypobaric hypoxia. Chronic hypoxic exposure caused no detectable change in eNOS protein levels in the lungs in either genotype; however, Mkp-1(-/-) mice had lower levels of eNOS protein and lower lung NO production than did WT mice. No iNOS protein was detected in the lungs by Western blotting in any condition in either genotype. Both arginase I and arginase II protein levels were greater in the lungs of hypoxic Mkp-1(-/-) mice than those in hypoxic WT mice. Lung levels of proliferating cell nuclear antigen were greater in hypoxic Mkp-1(-/-) than in hypoxic WT mice. These data are consistent with the concept that MKP-1 acts to restrain hypoxia-induced arginase expression and thereby reduces vascular remodeling and the severity of pulmonary hypertension.
Collapse
Affiliation(s)
- Yi Jin
- The Research Institute at Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Toby IT, Chicoine LG, Cui H, Chen B, Nelin LD. Hypoxia-induced proliferation of human pulmonary microvascular endothelial cells depends on epidermal growth factor receptor tyrosine kinase activation. Am J Physiol Lung Cell Mol Physiol 2010; 298:L600-6. [PMID: 20139181 DOI: 10.1152/ajplung.00122.2009] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We hypothesized that hypoxia would activate epidermal growth factor receptor (EGFR) tyrosine kinase, leading to increased arginase expression and resulting in proliferation of human pulmonary microvascular endothelial cell (hPMVEC). To test this hypothesis, hPMVEC were incubated in normoxia (20% O(2), 5% CO(2)) or hypoxia (1% O(2), 5% CO(2)). Immunoblotting for EGFR and proliferating cell nuclear antigen was done, and protein levels of both total EGFR and proliferating cell nuclear antigen were greater in hypoxic hPMVEC than in normoxic hPMVEC. Furthermore, hypoxic hPMVEC had greater levels of EGFR activity than did normoxic hPMVEC. Hypoxic hPMVEC had a twofold greater level of proliferation compared with normoxic controls, and this increase in proliferation was prevented by the addition of AG-1478 (a pharmacological inhibitor of EGFR). Immunoblotting for arginase I and arginase II demonstrated a threefold induction in arginase II protein levels in hypoxia, with little change in arginase I protein levels. The hypoxic induction of arginase II protein was prevented by treatment with AG-1478. Proliferation assays were performed in the presence of arginase inhibitors, and hypoxia-induced proliferation was also prevented by arginase inhibition. Finally, treatment with an EGFR small interfering RNA prevented hypoxia-induced proliferation and urea production. These findings demonstrate that hypoxia activates EGFR tyrosine kinase, leading to arginase expression and thereby promoting proliferation in hPMVEC.
Collapse
Affiliation(s)
- Inimary T Toby
- Pulmonary Hypertension Group, Center for Perinatal Research, Research Institute at Nationwide Children's Hospital, and Department of Pediatrics, Ohio State University, Columbus, Ohio, USA
| | | | | | | | | |
Collapse
|
29
|
Moss MB, Siqueira MA, Mann GE, Brunini TMC, Mendes-Ribeiro AC. Platelet aggregation in arterial hypertension: Is there a nitric oxide-urea connection? Clin Exp Pharmacol Physiol 2010; 37:167-72. [DOI: 10.1111/j.1440-1681.2009.05247.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
30
|
Sughrue ME, Yang I, Kane AJ, Rutkowski MJ, Fang S, James CD, Parsa AT. Immunological considerations of modern animal models of malignant primary brain tumors. J Transl Med 2009; 7:84. [PMID: 19814820 PMCID: PMC2768693 DOI: 10.1186/1479-5876-7-84] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2009] [Accepted: 10/08/2009] [Indexed: 12/26/2022] Open
Abstract
Recent advances in animal models of glioma have facilitated a better understanding of biological mechanisms underlying gliomagenesis and glioma progression. The limitations of existing therapy, including surgery, chemotherapy, and radiotherapy, have prompted numerous investigators to search for new therapeutic approaches to improve quantity and quality of survival from these aggressive lesions. One of these approaches involves triggering a tumor specific immune response. However, a difficulty in this approach is the the scarcity of animal models of primary CNS neoplasms which faithfully recapitulate these tumors and their interaction with the host's immune system. In this article, we review the existing methods utilized to date for modeling gliomas in rodents, with a focus on the known as well as potential immunological aspects of these models. As this review demonstrates, many of these models have inherent immune system limitations, and the impact of these limitations on studies on the influence of pre-clinical therapeutics testing warrants further attention.
Collapse
Affiliation(s)
- Michael E Sughrue
- Department of Neurological Surgery, University of California at San Francisco, San Francisco, California, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Chen B, Calvert AE, Cui H, Nelin LD. Hypoxia promotes human pulmonary artery smooth muscle cell proliferation through induction of arginase. Am J Physiol Lung Cell Mol Physiol 2009; 297:L1151-9. [PMID: 19801451 DOI: 10.1152/ajplung.00183.2009] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Vascular remodeling and smooth muscle cell proliferation are hallmark pathogenic features of pulmonary artery hypertension (PAH). Alterations in the metabolism of l-arginine via arginase and nitric oxide synthase play a critical role in the endothelial dysfunction seen in PAH. l-arginine metabolism by arginase produces l-ornithine and urea. l-ornithine is a precursor for polyamine and proline synthesis, ultimately leading to an increase in cellular proliferation. Given the integral role of the smooth muscle layer in the pathogenesis of hypoxia-induced PAH, we hypothesized that hypoxia would increase cellular proliferation via arginase induction in human pulmonary artery smooth muscle cells (hPASMC). We found that arginase II mRNA and protein expression were significantly increased in cultured hPASMC exposed to 1% O(2) for 24 and 48 h, which coincided with an increase in arginase activity at 48 h. There were no hypoxia-induced changes in levels of arginase I mRNA or protein in cultured hPASMC. Exposure to hypoxia resulted in more than one and a half times as many viable cells after 120 h than normoxic exposure. The addition of the arginase inhibitor, S-(2-boronoethyl)-l-cysteine, completely prevented both the hypoxia-induced increase in arginase activity and proliferation in hPASMC. Furthermore, transfection of small interfering RNA (siRNA) targeting arginase II in hPASMC resulted in knockdown of arginase II protein levels and complete prevention of the hypoxia-induced cellular proliferation. These data support our hypothesis that hypoxia increases proliferation of hPASMC through the induction of arginase II.
Collapse
Affiliation(s)
- Bernadette Chen
- Center for Perinatal Research, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, The Ohio State University College of Medicine, Columbus, USA.
| | | | | | | |
Collapse
|
32
|
Maarsingh H, Zaagsma J, Meurs H. Arginase: a key enzyme in the pathophysiology of allergic asthma opening novel therapeutic perspectives. Br J Pharmacol 2009; 158:652-64. [PMID: 19703164 DOI: 10.1111/j.1476-5381.2009.00374.x] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Allergic asthma is a chronic inflammatory airways' disease, characterized by allergen-induced early and late bronchial obstructive reactions, airway hyperresponsiveness (AHR), airway inflammation and airway remodelling. Recent ex vivo and in vivo studies in animal models and asthmatic patients have indicated that arginase may play a central role in all these features. Thus, increased arginase activity in the airways induces reduced bioavailability of L-arginine to constitutive (cNOS) and inducible (iNOS) nitric oxide synthases, causing a deficiency of bronchodilating and anti-inflammatory NO, as well as increased formation of peroxynitrite, which may be involved in allergen-induced airways obstruction, AHR and inflammation. In addition, both via reduced NO production and enhanced synthesis of L-ornithine, increased arginase activity may be involved in airway remodelling by promoting cell proliferation and collagen deposition in the airway wall. Therefore, arginase inhibitors may have therapeutic potential in the treatment of acute and chronic asthma. This review focuses on the pathophysiological role of arginase in allergic asthma and the emerging effectiveness of arginase inhibitors in the treatment of this disease.
Collapse
Affiliation(s)
- Harm Maarsingh
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.
| | | | | |
Collapse
|
33
|
Mabalirajan U, Aich J, Agrawal A, Ghosh B. Mepacrine inhibits subepithelial fibrosis by reducing the expression of arginase and TGF-beta1 in an extended subacute mouse model of allergic asthma. Am J Physiol Lung Cell Mol Physiol 2009; 297:L411-9. [PMID: 19542246 DOI: 10.1152/ajplung.00138.2009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Asthma is a dynamic disorder of airway inflammation and airway remodeling with an imbalance in T helper type 1 (Th(1))/Th(2) immune response. Increased Th(2) cytokines such as IL-4 and IL-13 induce arginase either directly or indirectly through transforming growth factor-beta(1) (TGF-beta(1)) and lead to subepithelial fibrosis, which is a crucial component of airway remodeling. Synthetic antimalarials have been reported to have immunomodulatory properties. Mepacrine is known for its reduction of airway inflammation in short-term allergen challenge model by reducing Th(2) cytokines and cysteinyl leukotrienes, which has an important role in the development of airway remodeling features. Therefore, we hypothesized that mepacrine may reduce airway remodeling. For this, extended subacute ovalbumin mice model of asthma was developed; these mice showed an increased expression of profibrotic mediators, subepithelial fibrosis, and goblet cell metaplasia along with airway inflammation, increased Th(2) cytokines, allergen-specific IgE, IgG(1), increased cytosolic PLA(2) (cPLA(2)), and airway hyperresponsiveness. Presence of intraepithelial eosinophils and significant TGF-beta(1) expression in subepithelial mesenchymal regions by repeated allergen exposures indicate that asthmatic mice of this study have developed human mimicking as well as late stages of asthma. However, mepacrine treatment decreased Th(2) cytokines and subepithelial fibrosis and alleviated asthma features. These reductions by mepacrine were associated with a decrease in levels and expression of TGF-beta(1) and the reduction in activity, expression of arginase in lung cytosol, and immunolocalization in inflammatory cells present in perivascular and peribronchial regions. These results suggest that mepacrine might reduce the development of subepithelial fibrosis by reducing the arginase and TGF-beta(1). These effects of mepacrine likely underlie its antiairway remodeling action in asthma.
Collapse
Affiliation(s)
- Ulaganathan Mabalirajan
- Molecular Immunogenetics Laboratory, Institute of Genomics and Integrative Biology, Mall Road, Delhi 110007, India
| | | | | | | |
Collapse
|
34
|
Morris SM. Recent advances in arginine metabolism: roles and regulation of the arginases. Br J Pharmacol 2009; 157:922-30. [PMID: 19508396 DOI: 10.1111/j.1476-5381.2009.00278.x] [Citation(s) in RCA: 344] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
As arginine can serve as precursor to a wide range of compounds, including nitric oxide, creatine, urea, polyamines, proline, glutamate and agmatine, there is considerable interest in elucidating mechanisms underlying regulation of its metabolism. It is now becoming apparent that the two isoforms of arginase in mammals play key roles in regulation of most aspects of arginine metabolism in health and disease. In particular, work over the past several years has focused on the roles and regulation of the arginases in vascular disease, pulmonary disease, infectious disease, immune cell function and cancer. As most of these topics have been considered in recent review articles, this review will focus more closely on results of recent studies on expression of the arginases in endothelial and vascular smooth muscle cells, post-translational modulation of arginase activity and applications of arginase inhibitors in vivo.
Collapse
Affiliation(s)
- Sidney M Morris
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA 15261, USA.
| |
Collapse
|
35
|
Ionova IA, Vásquez-Vivar J, Whitsett J, Herrnreiter A, Medhora M, Cooley BC, Pieper GM. Deficient BH4 production via de novo and salvage pathways regulates NO responses to cytokines in adult cardiac myocytes. Am J Physiol Heart Circ Physiol 2008; 295:H2178-87. [PMID: 18835915 PMCID: PMC2614582 DOI: 10.1152/ajpheart.00748.2008] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Accepted: 09/25/2008] [Indexed: 12/25/2022]
Abstract
Adult rat cardiac myocytes typically display a phenotypic response to cytokines manifested by low or no increases in nitric oxide (NO) production via inducible NO synthase (iNOS) that distinguishes them from other cell types. To better characterize this response, we examined the expression of tetrahydrobiopterin (BH4)-synthesizing and arginine-utilizing genes in cytokine-stimulated adult cardiac myocytes. Intracellular BH4 and 7,8-dihydrobiopterin (BH2) and NO production were quantified. Cytokines induced GTP cyclohydrolase and its feedback regulatory protein but with deficient levels of BH4 synthesis. Despite the induction of iNOS protein, cytokine-stimulated adult cardiac myocytes produced little or no increase in NO versus unstimulated cells. Western blot analysis under nonreducing conditions revealed the presence of iNOS monomers. Supplementation with sepiapterin (a precursor of BH4) increased BH4 as well as BH2, but this did not enhance NO levels or eliminate iNOS monomers. Similar findings were confirmed in vivo after treatment of rat cardiac allograft recipients with sepiapterin. It was found that expression of dihydrofolate reductase, required for full activity of the salvage pathway, was not detected in adult cardiac myocytes. Thus, adult cardiac myocytes have a limited capacity to synthesize BH4 after cytokine stimulation. The mechanisms involve posttranslational factors impairing de novo and salvage pathways. These conditions are unable to support active iNOS protein dimers necessary for NO production. These findings raise significant new questions about the prevailing understanding of how cytokines, via iNOS, cause cardiac dysfunction and injury in vivo during cardiac inflammatory disease states since cardiac myocytes are not a major source of high NO production.
Collapse
Affiliation(s)
- Irina A Ionova
- Department of Surgery (Transplant Surgery), Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Arginase and pulmonary diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2008; 378:171-84. [PMID: 18437360 PMCID: PMC2493601 DOI: 10.1007/s00210-008-0286-7] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Accepted: 03/17/2008] [Indexed: 10/31/2022]
Abstract
Recent studies have indicated that arginase, which converts L-arginine into L-ornithine and urea, may play an important role in the pathogenesis of various pulmonary disorders. In asthma, chronic obstructive pulmonary disease (COPD) and cystic fibrosis, increased arginase activity in the airways may contribute to obstruction and hyperresponsiveness of the airways by inducing a reduction in the production of bronchodilatory nitric oxide (NO) that results from its competition with constitutive (cNOS) and inducible (iNOS) NO synthases for their common substrate. In addition, reduced L-arginine availability to iNOS induced by arginase may result in the synthesis of both NO and the superoxide anion by this enzyme, thereby enhancing the production of peroxynitrite, which has procontractile and pro-inflammatory actions. Increased synthesis of L-ornithine by arginase may also contribute to airway remodelling in these diseases. L-Ornithine is a precursor of polyamines and L-proline, and these metabolic products may promote cell proliferation and collagen production, respectively. Increased arginase activity may also be involved in other fibrotic disorders of the lung, including idiopathic pulmonary fibrosis. Finally, through its action of inducing reduced levels of vasodilating NO, increased arginase activity has been associated with primary and secondary forms of pulmonary hypertension. Drugs targeting the arginase pathway could have therapeutic potential in these diseases.
Collapse
|
37
|
Maarsingh H, Zaagsma J, Meurs H. Arginine homeostasis in allergic asthma. Eur J Pharmacol 2008; 585:375-84. [PMID: 18410920 DOI: 10.1016/j.ejphar.2008.02.096] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2008] [Revised: 02/15/2008] [Accepted: 02/20/2008] [Indexed: 11/16/2022]
Abstract
Allergic asthma is a chronic disease characterized by early and late asthmatic reactions, airway hyperresponsiveness, airway inflammation and airway remodelling. Changes in l-arginine homeostasis may contribute to all these features of asthma by decreased nitric oxide (NO) production and increased formation of peroxynitrite, polyamines and l-proline. Intracellular l-arginine levels are regulated by at least three distinct mechanisms: (i) cellular uptake by cationic amino acid (CAT) transporters, (ii) metabolism by NO-synthase (NOS) and arginase, and (iii) recycling from l-citrulline. Ex vivo studies using animal models of allergic asthma have indicated that attenuated l-arginine bioavailability to NOS causes deficiency of bronchodilating NO and increased production of procontractile peroxynitrite, which importantly contribute to allergen-induced airway hyperresponsiveness after the early and late asthmatic reaction, respectively. Decreased cellular uptake of l-arginine, due to (eosinophil-derived) polycations inhibiting CATs, as well as increased consumption by increased arginase activity are major causes of substrate limitation to NOS. Increasing substrate availability to NOS by administration of l-arginine, l-citrulline, the polycation scavenger heparin, or an arginase inhibitor alleviates allergen-induced airway hyperresponsiveness by restoring the production of bronchodilating NO. In addition, reduced l-arginine levels may contribute to the airway inflammation associated with the development of airway hyperresponsiveness, which similarly may involve decreased NO synthesis and increased peroxynitrite formation. Increased arginase activity could also contribute to airway remodelling and persistent airway hyperresponsiveness in chronic asthma via increased synthesis of l-ornithine, the precursor of polyamines and l-proline. Drugs that increase the bioavailability of l-arginine in the airways - particularly arginase inhibitors - may have therapeutic potential in allergic asthma.
Collapse
Affiliation(s)
- Harm Maarsingh
- Department of Molecular Pharmacology, University Centre for Pharmacy, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | | | | |
Collapse
|
38
|
Durante W, Johnson FK, Johnson RA. Arginase: a critical regulator of nitric oxide synthesis and vascular function. Clin Exp Pharmacol Physiol 2007; 34:906-11. [PMID: 17645639 PMCID: PMC1955221 DOI: 10.1111/j.1440-1681.2007.04638.x] [Citation(s) in RCA: 411] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
1. Arginase is the focal enzyme of the urea cycle hydrolysing L-arginine to urea and L-ornithine. Emerging studies have identified arginase in the vasculature and have implicated this enzyme in the regulation of nitric oxide (NO) synthesis and the development of vascular disease. 2. Arginase inhibits the production of NO via several potential mechanisms, including competition with NO synthase (NOS) for the substrate L-arginine, uncoupling of NOS resulting in the generation of the NO scavenger, superoxide and peroxynitrite, repression of the translation and stability of inducible NOS protein, inhibition of inducible NOS activity via the generation of urea and by sensitization of NOS to its endogenous inhibitor asymmetric dimethyl-L-arginine. 3. Upregulation of arginase inhibits endothelial NOS-mediated NO synthesis and may contribute to endothelial dysfunction in hypertension, ageing, ischaemia-reperfusion and diabetes. 4. Arginase also redirects the metabolism of L-arginine to L-ornithine and the formation of polyamines and L-proline, which are essential for smooth muscle cell growth and collagen synthesis. Therefore, the induction of arginase may also promote aberrant vessel wall remodelling and neointima formation. 5. Arginase represents a promising novel therapeutic target that may reverse endothelial and smooth muscle cell dysfunction and prevent vascular disease.
Collapse
Affiliation(s)
- William Durante
- Department of Medical Pharmacology and Physiology, University of Missouri-Columbia, Columbia, Missouri 65212, USA.
| | | | | |
Collapse
|
39
|
Nelin LD, Wang X, Zhao Q, Chicoine LG, Young TL, Hatch DM, English BK, Liu Y. MKP-1 switches arginine metabolism from nitric oxide synthase to arginase following endotoxin challenge. Am J Physiol Cell Physiol 2007; 293:C632-40. [PMID: 17442735 DOI: 10.1152/ajpcell.00137.2006] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
L-Arginine (L-arg) is metabolized to nitric oxide (NO) by inducible NO synthase (iNOS) or to urea and L-ornithine (L-orn) by arginase. NO is involved in the inflammatory response, whereas arginase is the first step in polyamine and proline synthesis necessary for tissue repair and wound healing. Mitogen-activated protein kinases (MAPK) mediate LPS-induced iNOS expression, and MAPK phosphatase-1 (MKP-1) plays a crucial role in limiting MAPK signaling in macrophages. We hypothesized that MKP-1, by attenuating iNOS expression, acts as a switch changing L-arg metabolism from NO production to L-orn production after endotoxin administration. To test this hypothesis, we performed studies in RAW264.7 macrophages stably transfected with an MKP-1 expression vector in thioglyollate-elicited peritoneal macrophages harvested from wild-type and Mkp-1(-/-) mice, as well as in vivo in wild-type and Mkp-1(-/-) mice. We found that overexpression of MKP-1 resulted in lower iNOS expression and NO production but greater urea production in response to LPS. Although deficiency of MKP-1 resulted in greater iNOS expression and NO production and lower urea production in response to LPS, neither the overexpression nor the deficiency of MKP-1 had any substantial effect on the expression of the arginases.
Collapse
Affiliation(s)
- Leif D Nelin
- Center for Perinatal Research, Columbus Children's Research Institute, Columbus, OH 43205, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Demougeot C, Prigent-Tessier A, Bagnost T, André C, Guillaume Y, Bouhaddi M, Marie C, Berthelot A. Time course of vascular arginase expression and activity in spontaneously hypertensive rats. Life Sci 2006; 80:1128-34. [PMID: 17223136 DOI: 10.1016/j.lfs.2006.12.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2006] [Revised: 11/14/2006] [Accepted: 12/03/2006] [Indexed: 10/23/2022]
Abstract
There is growing evidence that vascular arginase plays a role in pathophysiology of vascular diseases. We recently reported high arginase activity/expression in young adult hypertensive spontaneously hypertensive rats (SHR). The aim of the present study was to characterize the time course of arginase pathway abnormalities in SHR and to explore the contributing role of hemodynamics and inflammation. Experiments were conducted on 5, 10, 19 and 26-week-old SHR and their age-matched control Wistar Kyoto (WKY) rats. Arginase activity as well as expression of arginase I, arginase II, endothelial and inducible NOS were determined in aortic tissue extracts. Levels of L-arginine, NO catabolites and IL-6 (a marker of inflammation) were measured in plasma. Arginase activity/expression was also measured in 10-week-old SHR previously treated with hydralazine (20 mg/kg/day, per os, for 5 weeks). As compared to WKY, SHR exhibited high vascular arginase I and II expression from prehypertensive to established stages of hypertension. However, a mismatch between expression and activity was observed at the prehypertensive stage. Arginase expression was not related either to plasma IL-6 levels or to expression of NOS. Prevention of hypertension by hydralazine significantly blunted arginase upregulation and restored arginase activity. Importantly, arginase activity and blood pressure (BP) correlated in SHR. In conclusion, our results demonstrate that arginase upregulation precedes blood pressure rising and identify elevated blood pressure as a contributing factor of arginase dysregulation in genetic hypertension. They also demonstrated a close relationship between arginase activity and BP, thus making arginase a promising target for antihypertensive therapy.
Collapse
Affiliation(s)
- Céline Demougeot
- Laboratoire de Physiologie, Pharmacologie, Nutrition Préventive Expérimentale, Equipe Optimisation Métabolique et Cellulaire, 25030 Besancon, France.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Decreased endothelial nitric oxide (NO) bioavailability as it relates to endothelial dysfunction plays an important role in various cardiovascular disorders, including athero-sclerosis. Recent research has provided evidence that endothelial dysfunction in atherosclerosis is not primarily caused by decreased endothelial NO synthase (eNOS) gene expression, but rather deregulation of eNOS enzymatic activity, which contributes to the increased oxidative stress in atherosclerosis. Among other mechanisms, the substrate L-arginine is an important limiting factor for NO production. Emerging evidence demonstrates that L-arginine is not only converted to NO via eNOS, but also metabolized to urea and l-ornithine via arginase in endothelial cells. Hence, arginase competes with eNOS for the substrate L-arginine, resulting in deceased NO production. There are an increasing number of studies showing that enhanced arginase gene expression and/or activity contribute to endothelial dysfunction in various cardiovascular disorders, including atherosclerosis. Thus, endothelial arginase may represent a new therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- Zhihong Yang
- Vascular Biology, Department of Medicine, Division of Physiology, University of Fribourg, Rue du Musée 5, CH-1700 Fribourg, Switzerland.
| | | |
Collapse
|