1
|
Molaro MC, Battisegola C, Schiano ME, Failla M, Rimoli MG, Lazzarato L, Chegaev K, Sodano F. Synthesis of Arginase Inhibitors: An Overview. Pharmaceutics 2025; 17:117. [PMID: 39861764 DOI: 10.3390/pharmaceutics17010117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/12/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Arginase (ARG) is a binuclear manganese-containing metalloenzyme that can convert L-arginine to L-ornithine and urea and plays a key role in the urea cycle. It also mediates different cellular functions and processes such as proliferation, senescence, apoptosis, autophagy, and inflammatory responses in various cell types. In mammals, there are two isoenzymes, ARG-1 and ARG-2; they are functionally similar, but their coding genes, tissue distribution, subcellular localization, and molecular regulation are distinct. In recent decades, the abnormal expression of ARG-1 or ARG-2 has been reported to be increasingly linked to a variety of diseases, including cardiovascular disease, inflammatory bowel disease, Alzheimer's disease, and cancer. Therefore, considering the current relevance of this topic and the need to address the growing demand for new and more potent ARG inhibitors in the context of various diseases, this review was conceived. We will provide an overview of all classes of ARG inhibitors developed so far including compounds of synthetic, natural, and semisynthetic origin. For the first time, the synthesis protocol and optimized reaction conditions of each molecule, including those reported in patent applications, will be described. For each molecule, its inhibitory activity in terms of IC50 towards ARG-1 and ARG-2 will be reported specifying the type of assay conducted.
Collapse
Affiliation(s)
| | - Chiara Battisegola
- Department of Pharmacy, "Federico II" University of Naples, 80131 Naples, Italy
| | | | - Mariacristina Failla
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Maria Grazia Rimoli
- Department of Pharmacy, "Federico II" University of Naples, 80131 Naples, Italy
| | - Loretta Lazzarato
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Konstantin Chegaev
- Department of Drug Science and Technology, University of Turin, 10125 Turin, Italy
| | - Federica Sodano
- Department of Pharmacy, "Federico II" University of Naples, 80131 Naples, Italy
| |
Collapse
|
2
|
Licata A, Seidita A, Como S, de Carlo G, Cammilleri M, Bonica R, Soresi M, Veronese N, Chianetta R, Citarrella R, Giannitrapani L, Barbagallo M. Herbal and Dietary Supplements as Adjunctive Treatment for Mild SARS-CoV-2 Infection in Italy. Nutrients 2025; 17:230. [PMID: 39861359 PMCID: PMC11767322 DOI: 10.3390/nu17020230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
During the COVID-19 pandemic, several observational studies proved a certain efficacy of nutraceuticals, herbal products, and other dietary supplements as adjuvant therapies used alongside antiviral drugs. Although their use has not been widespread in Italy, according to preliminary evidence, many supplements with demonstrated immunomodulatory effects, such as vitamins C and D, herbal medicines and essential oils, might relieve the respiratory symptoms of COVID-19, since SARS-CoV-2 can activate inflammasome-mediated inflammatory signaling pathways. Other observational studies have shown that herbal treatments, such as Echinacea purpurea and ginseng, help alleviate respiratory symptoms and reduce serum levels of inflammatory cytokines, which are typically overexpressed in both adult and pediatric SARS-CoV-2 patients. Further, vitamins C and D can attenuate the immune response thanks to their cytokine suppression ability and to their known antimicrobial activity and potential to modulate T helper cell response. The strong immune response triggered by SARS-CoV-2 infection is responsible for the severity of the disease. Preliminary data have also shown that L-arginine, an endothelial-derived relaxing factor, is able to modulate endothelial damage, which appears to be one of the main targets of this systemic disease. Finally, some essential oils and their isolated compounds, such as eucalyptol, may be helpful in reducing many of the respiratory symptoms of COVID-19, although others, such as menthol, are not recommended, since it can lead to an undervaluation of the clinical status of a patient. In this narrative review, despite the lack of strong evidence in this field, we aimed to give an overview of the current available literature (mainly observational and cross-sectional studies) regarding herbal products and dietary supplements and their use in the treatment of mild disease from SARS-CoV-2 infection. Obviously, dietary supplements and herbal products do not constitute a standardized treatment for COVID-19 disease, but they could represent an adjunctive and useful treatment when used together with antivirals.
Collapse
Affiliation(s)
- Anna Licata
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Aurelio Seidita
- Unit of Internal Medicine, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, 90146 Palermo, Italy; (A.S.)
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90146 Palermo, Italy
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Silvia Como
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Gabriele de Carlo
- Unit of Internal Medicine, “V. Cervello” Hospital, Ospedali Riuniti “Villa Sofia-Cervello”, 90146 Palermo, Italy; (A.S.)
| | - Marcella Cammilleri
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Roberta Bonica
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Maurizio Soresi
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Nicola Veronese
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Roberta Chianetta
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Roberto Citarrella
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| | - Lydia Giannitrapani
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), 90146 Palermo, Italy
| | - Mario Barbagallo
- Unit of Internal Medicine, AOU Policlinico “P. Giaccone”, Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90134 Palermo, Italy; (S.C.); (M.C.); (N.V.)
| |
Collapse
|
3
|
Wu S, Zhao W, Yu Z. Novel Targets and Potential Mechanisms of Mizuhopecten yessoensis-Derived Tripeptide NCW as Antihypertensive Peptides. Mol Nutr Food Res 2024; 68:e2300552. [PMID: 38366946 DOI: 10.1002/mnfr.202300552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/07/2023] [Indexed: 02/19/2024]
Abstract
SCOPE Mizuhopecten yessoensis-derived tripeptide Asn-Cys-Trp (NCW) exhibits a potent antihypertensive effect in vivo. However, a lack of knowledge of the antihypertensive mechanism of tripeptide NCW limits its application for functional foods industrialization. The purpose of this study is to elucidate the corresponding targets and mechanisms of tripeptide NCW in hypertension regulation. METHODS AND RESULTS Administration of tripeptide NCW for 3 weeks, the blood pressure of spontaneously hypertensive rats (SHRs) is significantly decreased. After sacrifice, the serum sample is analyzed using tandem mass tag (TMT)-based liquid chromatography with tandem mass spectrometry to identify differentially expressed proteins. The proteomic analysis indicates that tripeptide NCW administration alters serum protein profiles in SHR rats, significantly upregulating 106 proteins and downregulating 30 proteins. These proteins enhance the glycolysis, glucose, and TCA cycle, improve amino metabolism, trigger the cAMP/PKA, cGMP/PKG, PI3K/AKT, and AMPK signal pathways, and inhibit Ras-regulated JNK activation, TGF-β/MAPK, and TGF-β/ RhoA/ROCK pathways. CONCLUSION Tripeptide NCW supplementation is demonstrated to regulate signal pathways involved in the control of blood pressure and regulate the energy and amino acids metabolic processes in serum, providing important insights into the protective effects of tripeptide NCW on hypertension.
Collapse
Affiliation(s)
- Sijia Wu
- School of Food Science and Engineering, Hainan University, Haikou, 570228, P.R. China
- College of Food Science and Engineering, Jilin University, Changchun, 130062, P.R. China
| | - Wenzhu Zhao
- School of Food Science and Engineering, Hainan University, Haikou, 570228, P.R. China
| | - Zhipeng Yu
- School of Food Science and Engineering, Hainan University, Haikou, 570228, P.R. China
| |
Collapse
|
4
|
Toso A, Aránguiz O, Céspedes C, Navarrete O, Hernández C, Vio CP, Luco M, Casanello P, Kattan J. Congenital diaphragmatic hernia: phosphodiesterase-5 and Arginase inhibitors prevent pulmonary vascular hypoplasia in rat lungs. Pediatr Res 2024; 95:941-948. [PMID: 36418485 DOI: 10.1038/s41390-022-02366-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/24/2022] [Accepted: 09/28/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Severe pulmonary hypoplasia related to congenital diaphragmatic hernia (CDH) continues to be a potentially fatal condition despite advanced postnatal management strategies. OBJECTIVE To evaluate the effect of the antenatal sildenafil and 2(S)-amino-6-boronohexanoic acid (ABH-Arginase inhibitor) on lung volume, pulmonary vascular development, and nitric oxide (NO) synthesis in a Nitrofen-induced CDH rat model. METHODS Nitrofen-induced CDH rat model was used. Nitrofen was administrated on embryonic day(E) 9,5. At E14, five intervention groups were treated separately: Nitrofen, Nitrofen+Sildenafil, Nitrofen+ABH, Nitrofen+Sildenafil+ABH and Control. At term, offspring's lungs were weighed, some paraffin-embedded for histology, others snap-frozen to analyze eNOS, Arginase I-II expression, and activity. RESULTS In CDH-bearing offsprings, ABH or Sildenafil+ABH preserved the total lung/body-weight index (p < 0.001), preventing pulmonary vascular smooth muscle cell hyperproliferation and improving lung morphometry. Sildenafil+ABH increased 1.7-fold the lung nitrite levels (p < 0.01) without changes in eNOS expression. Sildenafil and ABH improved the number of pulmonary vessels. CONCLUSION These results suggest that in this CDH rat model, the basal activity of Arginase participates in the lung volume and, together with phosphodiesterase-5, regulates NOS activity in the term fetal lung. The combined treatment (Sildenafil+ABH) could revert some of the pulmonary features in CDH by improving the local NO synthesis and preventing smooth muscle cell hyperproliferation. IMPACT This study presents Arginase inhibition as a new therapeutic target and the importance of the combined antenatal treatment to improve pulmonary vascular development in a congenital diaphragmatic hernia (CDH) rat model. This study shows that the action of an Arginase inhibitor (ABH) enhances the effects already described for sildenafil in this model. These results reinforce the importance of prenatal treatments' synergy in recovering the hypoplastic lung in the Nitrofen-induced CDH rat model.
Collapse
Affiliation(s)
- Alberto Toso
- Department of Neonatology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Oscar Aránguiz
- Department of Neonatology, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Obstetrics, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos Céspedes
- Center for Aging and Regeneration CARE UC, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| | - Orieta Navarrete
- Department of Pathology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cherie Hernández
- Department of Neonatology, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Obstetrics, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos P Vio
- Center for Aging and Regeneration CARE UC, Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
- Faculty of Medicine and Science, Universidad San Sebastián, Santiago, Chile
| | - Matías Luco
- Department of Neonatology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Paola Casanello
- Department of Neonatology, Pontificia Universidad Católica de Chile, Santiago, Chile.
- Department of Obstetrics, Pontificia Universidad Católica de Chile, Santiago, Chile.
| | - Javier Kattan
- Department of Neonatology, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
5
|
Wang Y, Wang M, Wang Y. Irisin: A Potentially Fresh Insight into the Molecular Mechanisms Underlying Vascular Aging. Aging Dis 2023; 15:2491-2506. [PMID: 38029393 PMCID: PMC11567262 DOI: 10.14336/ad.2023.1112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 11/12/2023] [Indexed: 12/01/2023] Open
Abstract
Aging is a natural process that affects all living organisms, including humans. Aging is a complex process that involves the gradual deterioration of various biological processes and systems, including the cardiovascular system. Vascular aging refers to age-related changes in blood vessels. These changes can increase the risk of developing cardiovascular diseases, such as hypertension, atherosclerosis, and stroke. Recently, an exercise-induced muscle factor, irisin, was found to directly improve metabolism and regulate the balance of glucolipid metabolism, thereby counteracting obesity and insulin resistance. Based on a growing body of evidence, irisin modulates vascular aging. Adenosine monophosphate-activated protein kinase (AMPK) serves as a pivotal cellular energy sensor and metabolic modulator, acting as a central signaling cascade to coordinate various cellular processes necessary for maintaining vascular homeostasis. The vascular regulatory effects of irisin are closely intertwined with its interaction with the AMPK pathway. In conclusion, understanding the molecular processes used by irisin to regulate changes in vascular diseases caused by aging may inspire the development of techniques that promote healthy vascular aging. This review sought to describe the impact of irisin on the molecular mechanisms of vascular aging, including inflammation, oxidative stress, and epigenetics, from the perspective of endothelial cell function and vascular macroregulation, and summarize the multiple signaling pathways used by irisin to regulate vascular aging.
Collapse
Affiliation(s)
- Yinghui Wang
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, Jilin, China.
| | - Manying Wang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China.
| | - Yuehui Wang
- Department of Geriatrics, Jilin Geriatrics Clinical Research Center, The First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
6
|
Belinskaia DA, Voronina PA, Popova PI, Voitenko NG, Shmurak VI, Vovk MA, Baranova TI, Batalova AA, Korf EA, Avdonin PV, Jenkins RO, Goncharov NV. Albumin Is a Component of the Esterase Status of Human Blood Plasma. Int J Mol Sci 2023; 24:10383. [PMID: 37373530 PMCID: PMC10299176 DOI: 10.3390/ijms241210383] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
The esterase status of blood plasma can claim to be one of the universal markers of various diseases; therefore, it deserves attention when searching for markers of the severity of COVID-19 and other infectious and non-infectious pathologies. When analyzing the esterase status of blood plasma, the esterase activity of serum albumin, which is the major protein in the blood of mammals, should not be ignored. The purpose of this study is to expand understanding of the esterase status of blood plasma and to evaluate the relationship of the esterase status, which includes information on the amount and enzymatic activity of human serum albumin (HSA), with other biochemical parameters of human blood, using the example of surviving and deceased patients with confirmed COVID-19. In experiments in vitro and in silico, the activity of human plasma and pure HSA towards various substrates was studied, and the effect of various inhibitors on this activity was tested. Then, a comparative analysis of the esterase status and a number of basic biochemical parameters of the blood plasma of healthy subjects and patients with confirmed COVID-19 was performed. Statistically significant differences have been found in esterase status and biochemical indices (including albumin levels) between healthy subjects and patients with COVID-19, as well as between surviving and deceased patients. Additional evidence has been obtained for the importance of albumin as a diagnostic marker. Of particular interest is a new index, [Urea] × [MDA] × 1000/(BChEb × [ALB]), which in the group of deceased patients was 10 times higher than in the group of survivors and 26 times higher than the value in the group of apparently healthy elderly subjects.
Collapse
Affiliation(s)
- Daria A. Belinskaia
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, pr. Torez 44, 194223 St. Petersburg, Russia
| | - Polina A. Voronina
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, pr. Torez 44, 194223 St. Petersburg, Russia
| | - Polina I. Popova
- City Polyclinic No. 112, 25 Academician Baykov Str., 195427 St. Petersburg, Russia
| | - Natalia G. Voitenko
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, pr. Torez 44, 194223 St. Petersburg, Russia
| | - Vladimir I. Shmurak
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, pr. Torez 44, 194223 St. Petersburg, Russia
| | - Mikhail A. Vovk
- Centre for Magnetic Resonance, St. Petersburg State University, Universitetskij pr., 26, Peterhof, 198504 St. Petersburg, Russia
| | - Tatiana I. Baranova
- Faculty of Biology, St. Petersburg State University, 7-9 Universitetskaya Emb., 199034 St. Petersburg, Russia
| | - Anastasia A. Batalova
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, pr. Torez 44, 194223 St. Petersburg, Russia
| | - Ekaterina A. Korf
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, pr. Torez 44, 194223 St. Petersburg, Russia
| | - Pavel V. Avdonin
- Koltsov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilova Str., 119334 Moscow, Russia
| | - Richard O. Jenkins
- Leicester School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | - Nikolay V. Goncharov
- Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, pr. Torez 44, 194223 St. Petersburg, Russia
| |
Collapse
|
7
|
Miklós Z, Horváth I. The Role of Oxidative Stress and Antioxidants in Cardiovascular Comorbidities in COPD. Antioxidants (Basel) 2023; 12:1196. [PMID: 37371927 DOI: 10.3390/antiox12061196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Oxidative stress driven by several environmental and local airway factors associated with chronic obstructive bronchiolitis, a hallmark feature of COPD, plays a crucial role in disease pathomechanisms. Unbalance between oxidants and antioxidant defense mechanisms amplifies the local inflammatory processes, worsens cardiovascular health, and contributes to COPD-related cardiovascular dysfunctions and mortality. The current review summarizes recent developments in our understanding of different mechanisms contributing to oxidative stress and its countermeasures, with special attention to those that link local and systemic processes. Major regulatory mechanisms orchestrating these pathways are also introduced, with some suggestions for further research in the field.
Collapse
Affiliation(s)
- Zsuzsanna Miklós
- National Korányi Institute for Pulmonology, Korányi F. Street 1, H-1121 Budapest, Hungary
| | - Ildikó Horváth
- National Korányi Institute for Pulmonology, Korányi F. Street 1, H-1121 Budapest, Hungary
- Department of Pulmonology, University of Debrecen, Nagyerdei krt 98, H-4032 Debrecen, Hungary
| |
Collapse
|
8
|
Douglass MS, Kaplowitz MR, Zhang Y, Fike CD. Impact of l-citrulline on nitric oxide signaling and arginase activity in hypoxic human pulmonary artery endothelial cells. Pulm Circ 2023; 13:e12221. [PMID: 37063746 PMCID: PMC10091859 DOI: 10.1002/pul2.12221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/18/2023] Open
Abstract
Impaired nitric oxide (NO) signaling contributes to the development of pulmonary hypertension (PH). The l-arginine precursor, l-citrulline, improves NO signaling and has therapeutic potential in PH. However, there is evidence that l-citrulline might increase arginase activity, which in turn, has been shown to contribute to PH. Our major purpose was to determine if l-citrulline increases arginase activity in hypoxic human pulmonary artery endothelial cells (PAECs). In addition, to avoid potential adverse effects from high dose l-citrulline monotherapy, we evaluated whether the effect on NO signaling is greater using co-treatment with l-citrulline and another agent that improves NO signaling, folic acid, than either alone. Arginase activity was measured in human PAECs cultured under hypoxic conditions in the presence of l-citrulline (0-1 mM). NO production and endothelial nitric oxide synthase (eNOS) coupling, as assessed by eNOS dimer-to-monomer ratios, were measured in PAECs treated with l-citrulline and/or folic acid (0.2 μM). Arginase activity increased in hypoxic PAECs treated with 1 mM but not with either 0.05 or 0.1 mM l-citrulline. Co-treatment with folic acid and 0.1 mM l-citrulline increased NO production and eNOS dimer-to-monomer ratios more than treatment with either alone. The potential to increase arginase activity suggests that there might be plasma l-citrulline concentrations that should not be exceeded when using l-citrulline to treat PH. Rather than progressively increasing the dose of l-citrulline as a monotherapy, co-therapy with l-citrulline and folic acid merits consideration, due to the possibility of achieving efficacy at lower doses and minimizing side effects.
Collapse
Affiliation(s)
| | | | - Yongmei Zhang
- Department of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| | - Candice D. Fike
- Department of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| |
Collapse
|
9
|
Serna E, Mauricio MD, San-Miguel T, Guerra-Ojeda S, Verdú D, Valls A, Arc-Chagnaud C, De la Rosa A, Viña J. Glucose 6-P Dehydrogenase Overexpression Improves Aging-Induced Endothelial Dysfunction in Aorta from Mice: Role of Arginase II. Int J Mol Sci 2023; 24:ijms24043622. [PMID: 36835034 PMCID: PMC9961129 DOI: 10.3390/ijms24043622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
The increase of vascular arginase activity during aging causes endothelial dysfunction. This enzyme competes with the endothelial nitric oxide synthase (eNOS) for L-arginine substrate. Our hypothesis is that glucose 6-P dehydrogenase (G6PD) overexpression could improve the endothelial function modulating the arginase pathway in aorta from mice. For this study, three groups of male mice were used: young wild type (WT) (6-9 months), old WT (21-22 months) and old G6PD-Tg (21-22 months) mice. Vascular reactivity results showed a reduced acetylcholine-dependent relaxation in the old WT but not old G6PD-Tg group. Endothelial dysfunction was reverted by nor-NOHA, an arginase inhibitor. Mice overexpressing G6PD underexpressed arginase II and also displayed a lower activity of this enzyme. Moreover, histological analyses demonstrated that age causes a thickness of aortic walls, but this did not occur in G6PD-Tg mice. We conclude that the overexpressing G6PD mouse is a model to improve vascular health via the arginase pathway.
Collapse
Affiliation(s)
- Eva Serna
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
- Correspondence:
| | - Maria D Mauricio
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Teresa San-Miguel
- Department of Pathology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Sol Guerra-Ojeda
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - David Verdú
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Alicia Valls
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Coralie Arc-Chagnaud
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Adrián De la Rosa
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - José Viña
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
10
|
Chen P, Liu X, Gu C, Zhong P, Song N, Li M, Dai Z, Fang X, Liu Z, Zhang J, Tang R, Fan S, Lin X. A plant-derived natural photosynthetic system for improving cell anabolism. Nature 2022; 612:546-554. [PMID: 36477541 PMCID: PMC9750875 DOI: 10.1038/s41586-022-05499-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 10/31/2022] [Indexed: 12/12/2022]
Abstract
Insufficient intracellular anabolism is a crucial factor involved in many pathological processes in the body1,2. The anabolism of intracellular substances requires the consumption of sufficient intracellular energy and the production of reducing equivalents. ATP acts as an 'energy currency' for biological processes in cells3,4, and the reduced form of NADPH is a key electron donor that provides reducing power for anabolism5. Under pathological conditions, it is difficult to correct impaired anabolism and to increase insufficient levels of ATP and NADPH to optimum concentrations1,4,6-8. Here we develop an independent and controllable nanosized plant-derived photosynthetic system based on nanothylakoid units (NTUs). To enable cross-species applications, we use a specific mature cell membrane (the chondrocyte membrane (CM)) for camouflage encapsulation. As proof of concept, we demonstrate that these CM-NTUs enter chondrocytes through membrane fusion, avoid lysosome degradation and achieve rapid penetration. Moreover, the CM-NTUs increase intracellular ATP and NADPH levels in situ following exposure to light and improve anabolism in degenerated chondrocytes. They can also systemically correct energy imbalance and restore cellular metabolism to improve cartilage homeostasis and protect against pathological progression of osteoarthritis. Our therapeutic strategy for degenerative diseases is based on a natural photosynthetic system that can controllably enhance cell anabolism by independently providing key energy and metabolic carriers. This study also provides an enhanced understanding of the preparation and application of bioorganisms and composite biomaterials for the treatment of disease.
Collapse
Affiliation(s)
- Pengfei Chen
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xin Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Chenhui Gu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Peiyu Zhong
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Nan Song
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Mobai Li
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhanqiu Dai
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Xiangqian Fang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Zhaoming Liu
- Department of Chemistry, Zhejiang University, Hangzhou, China
| | - Jianfeng Zhang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China
| | - Ruikang Tang
- Department of Chemistry, Zhejiang University, Hangzhou, China.
| | - Shunwu Fan
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| | - Xianfeng Lin
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
11
|
Yadav S, Priya A, Borade DR, Agrawal-Rajput R. Macrophage subsets and their role: co-relation with colony-stimulating factor-1 receptor and clinical relevance. Immunol Res 2022; 71:130-152. [PMID: 36266603 PMCID: PMC9589538 DOI: 10.1007/s12026-022-09330-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/14/2022] [Indexed: 01/10/2023]
Abstract
Macrophages are one of the first innate immune cells to reach the site of infection or injury. Diverse functions from the uptake of pathogen or antigen, its killing, and presentation, the release of pro- or anti-inflammatory cytokines, activation of adaptive immune cells, clearing off tissue debris, tissue repair, and maintenance of tissue homeostasis have been attributed to macrophages. Besides tissue-resident macrophages, the circulating macrophages are recruited to different tissues to get activated. These are highly plastic cells, showing a spectrum of phenotypes depending on the stimulus received from their immediate environment. The macrophage differentiation requires colony-stimulating factor-1 (CSF-1) or macrophage colony-stimulating factor (M-CSF), colony-stimulating factor-2 (CSF-2), or granulocyte–macrophage colony-stimulating factor (GM-CSF) and different stimuli activate them to different phenotypes. The richness of tissue macrophages is precisely controlled via the CSF-1 and CSF-1R axis. In this review, we have given an overview of macrophage origin via hematopoiesis/myelopoiesis, different phenotypes associated with macrophages, their clinical significance, and how they are altered in various diseases. We have specifically focused on the function of CSF-1/CSF-1R signaling in deciding macrophage fate and the outcome of aberrant CSF-1R signaling in relation to macrophage phenotype in different diseases. We further extend the review to briefly discuss the possible strategies to manipulate CSF-1R and its signaling with the recent updates.
Collapse
Affiliation(s)
- Shivani Yadav
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, 382426, Gujarat, India
| | - Astik Priya
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, 382426, Gujarat, India
| | - Diksha R Borade
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, 382426, Gujarat, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Indian Institute of Advanced Research, Gandhinagar, 382426, Gujarat, India.
| |
Collapse
|
12
|
Vanillic Acid Attenuates Monocrotaline-Induced Pulmonary Arterial Hypertension by Enhancing NO Signaling Pathways. Nat Prod Commun 2022. [DOI: 10.1177/1934578x221128411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a severe progressive disease characterized by elevated mean pulmonary arterial pressure, right ventricular hypertrophy, and eventual progression to right heart failure and death. This study aimed to examine the effect of the natural product vanillic acid (VA) on monocrotaline (MCT)-induced PAH in rats. The arginase inhibitory activity and enzyme kinetic reaction of VA were also investigated. The results showed that VA could improve pulmonary arterial pressure, pulmonary artery vascular remodeling, and right ventricular remodeling induced by MCT in rats and reduce the degree of pulmonary tissue fibrosis. Moreover, VA downregulated the gene and protein expression levels of Hif-2α, Hif-1β, and Arg2 and increased the P-eNOS/eNOS levels, thus increasing nitric oxide (NO) levels in PAH rats. Furthermore, VA was determined to be a mixed competitive arginase inhibitor with an IC50 of 26.1 μM. In conclusion, the arginase inhibitor VA exerted protective effects on MCT-induced PAH and pulmonary vascular remodeling by enhancing NO signaling pathways.
Collapse
|
13
|
Detroja TS, Samson AO. Virtual Screening for FDA-Approved Drugs That Selectively Inhibit Arginase Type 1 and 2. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27165134. [PMID: 36014374 PMCID: PMC9416497 DOI: 10.3390/molecules27165134] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/30/2022]
Abstract
Arginases are often overexpressed in human diseases, and they are an important target for developing anti-aging and antineoplastic drugs. Arginase type 1 (ARG1) is a cytosolic enzyme, and arginase type 2 (ARG2) is a mitochondrial one. In this study, a dataset containing 2115-FDA-approved drug molecules is virtually screened for potential arginase binding using molecular docking against several ARG1 and ARG2 structures. The potential arginase ligands are classified into three categories: (1) Non-selective, (2) ARG1 selective, and (3) ARG2 selective. The evaluated potential arginase ligands are then compared with their clinical use. Remarkably, half of the top 30 potential drugs are used clinically to lower blood pressure and treat cancer, infection, kidney disease, and Parkinson’s disease thus partially validating our virtual screen. Most notable are the antihypertensive drugs candesartan, irbesartan, indapamide, and amiloride, the antiemetic rolapitant, the anti-angina ivabradine, and the antidiabetic metformin which have minimal side effects. The partial validation also favors the idea that the other half of the top 30 potential drugs could be used in therapeutic settings. The three categories greatly expand the selectivity of arginase inhibition.
Collapse
|
14
|
Xiao-Chai-Hu Decoction Ameliorates Poly (I:C)-Induced Viral Pneumonia through Inhibiting Inflammatory Response and Modulating Serum Metabolism. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1240242. [PMID: 35865338 PMCID: PMC9296287 DOI: 10.1155/2022/1240242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/27/2022] [Accepted: 06/15/2022] [Indexed: 11/30/2022]
Abstract
Viral pneumonia is widespread, progresses rapidly, and has a high mortality rate. Developing safe and effective therapies to treat viral pneumonia can minimize risks to public health and alleviate pressures on the associated health systems. Xiao-Chai-Hu (XCH) decoction can be used in the treatment of viral pneumonia. However, the mechanisms of XCH on viral pneumonia remain unclear. In this study, poly (I:C) was used to establish a mouse model of viral pneumonia, and the therapeutic effects of XCH on viral pneumonia were assessed. Furthermore, we evaluated the effects of XCH on inflammatory response. Lastly, untargeted metabolomics were used to study the metabolic regulatory mechanisms of XCH on viral pneumonia model mice. Our results showed that XCH treatment decreased the wet/dry ratio in lung tissue, total protein concentration, and total cell count in bronchoalveolar lavage fluid (BALF). H&E staining indicated that XCH treatment alleviated the pathological changes in lung. Moreover, XCH treatment decreased the levels of proinflammatory cytokines (IL-1β, IL-6, and TNF-α) and lowered the ratio of CD86+/CD206+ macrophages and CD11b+LY6G+ neutrophils in BALF. XCH treatment also decreased the myeloperoxidase (MPO) and reduced the phosphorylations of PI3K, AKT, and NF-κB p65 in lung. Serum untargeted metabolomics analysis showed that XCH treatment could affect 18 metabolites in serum such as creatine, hydroxyproline, cortisone, hydrocortisone, corticosterone, hypotaurine, and taurine. These metabolites were associated with arginine and proline metabolism, steroid hormone biosynthesis, and taurine and hypotaurine metabolism processes. In conclusion, our study demonstrated that treatment with XCH can ameliorate viral pneumonia and reduce inflammatory response in viral pneumonia. The mechanism of action of XCH in the treatment of viral pneumonia may be associated with inhibiting the activation of PI3K/AKT/NF-κB signaling pathway in lung and regulating arginine and proline metabolism, steroid hormone biosynthesis, and taurine and hypotaurine metabolism in serum.
Collapse
|
15
|
Poznyak AV, Sadykhov NK, Kartuesov AG, Borisov EE, Sukhorukov VN, Orekhov AN. Aging of Vascular System Is a Complex Process: The Cornerstone Mechanisms. Int J Mol Sci 2022; 23:ijms23136926. [PMID: 35805936 PMCID: PMC9266404 DOI: 10.3390/ijms23136926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/15/2022] [Accepted: 06/20/2022] [Indexed: 02/04/2023] Open
Abstract
Aging is one of the most intriguing processes of human ontogenesis. It is associated with the development of a wide variety of diseases affecting all organs and their systems. The victory over aging is the most desired goal of scientists; however, it is hardly achievable in the foreseeable future due to the complexity and ambiguity of the process itself. All body systems age, lose their performance, and structural disorders accumulate. The cardiovascular system is no exception. And it is cardiovascular diseases that occupy a leading position as a cause of death, especially among the elderly. The aging of the cardiovascular system is well described from a mechanical point of view. Moreover, it is known that at the cellular level, a huge number of mechanisms are involved in this process, from mitochondrial dysfunction to inflammation. It is on these mechanisms, as well as the potential for taking control of the aging of the cardiovascular system, that we focused on in this review.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia;
- Correspondence: (A.V.P.); (A.N.O.)
| | - Nikolay K. Sadykhov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.K.S.); (A.G.K.)
| | - Andrey G. Kartuesov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.K.S.); (A.G.K.)
| | - Evgeny E. Borisov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky National Research Centre of Surgery, 2, Abrikosovsky Lane, 119991 Moscow, Russia;
| | - Vasily N. Sukhorukov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia;
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky National Research Centre of Surgery, 2, Abrikosovsky Lane, 119991 Moscow, Russia;
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Osennyaya 4-1-207, 121609 Moscow, Russia;
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, 125315 Moscow, Russia; (N.K.S.); (A.G.K.)
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky National Research Centre of Surgery, 2, Abrikosovsky Lane, 119991 Moscow, Russia;
- Correspondence: (A.V.P.); (A.N.O.)
| |
Collapse
|
16
|
Chao de la Barca JM, Richard A, Robert P, Eid M, Fouquet O, Tessier L, Wetterwald C, Faure J, Fassot C, Henrion D, Reynier P, Loufrani L. Metabolomic Profiling of Angiotensin-II-Induced Abdominal Aortic Aneurysm in Ldlr -/- Mice Points to Alteration of Nitric Oxide, Lipid, and Energy Metabolisms. Int J Mol Sci 2022; 23:ijms23126387. [PMID: 35742839 PMCID: PMC9223449 DOI: 10.3390/ijms23126387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/28/2022] [Accepted: 06/03/2022] [Indexed: 11/16/2022] Open
Abstract
Aneurysm is the second-most common disease affecting the aorta worldwide after atherosclerosis. While several clinical metabolomic studies have been reported, no study has reported deep metabolomic phenotyping in experimental animal models of aortic aneurysm. We performed a targeted metabolomics study on the blood and aortas of an experimental mice model of aortic aneurysm generated by high-cholesterol diet and angiotensin II in Ldlr−/− mice. The mice model showed a significant increase in media/lumen ratio and wall area, which is associated with lipid deposition within the adventitia, describing a hypertrophic remodeling with an aneurysm profile of the abdominal aorta. Altered aortas showed increased collagen remodeling, disruption of lipid metabolism, decreased glucose, nitric oxide and lysine metabolisms, and increased polyamines and asymmetric dimethylarginine (ADMA) production. In blood, a major hyperlipidemia was observed with decreased concentrations of glutamine, glycine, taurine, and carnitine, and increased concentrations of the branched amino acids (BCAA). The BCAA/glycine and BCAA/glutamine ratios discriminated with very good sensitivity and specificity between aneurysmatic and non-aneurysmatic mice. To conclude, our results reveal that experimental induction of aortic aneurysms causes a profound alteration in the metabolic profile in aortas and blood, mainly centered on an alteration of NO, lipid, and energetic metabolisms.
Collapse
Affiliation(s)
- Juan Manuel Chao de la Barca
- UMR CNRS 6015—INSERM U1083, IRIS2, 3 rue Roger Amsler, 49100 Angers, France; (J.M.C.d.l.B.); (A.R.); (P.R.); (C.F.); (D.H.); (P.R.)
- INSERM U1083, 49100 Angers, France
- Mitovasc Institute, Université d’Angers, 49100 Angers, France
- Service de Biochimie et Biologie Moléculaire, Centre Hospitalier Universitaire (CHU), 49000 Angers, France; (L.T.); (C.W.); (J.F.)
| | - Alexis Richard
- UMR CNRS 6015—INSERM U1083, IRIS2, 3 rue Roger Amsler, 49100 Angers, France; (J.M.C.d.l.B.); (A.R.); (P.R.); (C.F.); (D.H.); (P.R.)
- INSERM U1083, 49100 Angers, France
- Mitovasc Institute, Université d’Angers, 49100 Angers, France
| | - Pauline Robert
- UMR CNRS 6015—INSERM U1083, IRIS2, 3 rue Roger Amsler, 49100 Angers, France; (J.M.C.d.l.B.); (A.R.); (P.R.); (C.F.); (D.H.); (P.R.)
- INSERM U1083, 49100 Angers, France
- Mitovasc Institute, Université d’Angers, 49100 Angers, France
| | - Maroua Eid
- Service de Chirurgie Cardiaque, Centre Hospitalier Universitaire (CHU), 49100 Angers, France; (M.E.); (O.F.)
| | - Olivier Fouquet
- Service de Chirurgie Cardiaque, Centre Hospitalier Universitaire (CHU), 49100 Angers, France; (M.E.); (O.F.)
| | - Lydie Tessier
- Service de Biochimie et Biologie Moléculaire, Centre Hospitalier Universitaire (CHU), 49000 Angers, France; (L.T.); (C.W.); (J.F.)
| | - Céline Wetterwald
- Service de Biochimie et Biologie Moléculaire, Centre Hospitalier Universitaire (CHU), 49000 Angers, France; (L.T.); (C.W.); (J.F.)
| | - Justine Faure
- Service de Biochimie et Biologie Moléculaire, Centre Hospitalier Universitaire (CHU), 49000 Angers, France; (L.T.); (C.W.); (J.F.)
| | - Celine Fassot
- UMR CNRS 6015—INSERM U1083, IRIS2, 3 rue Roger Amsler, 49100 Angers, France; (J.M.C.d.l.B.); (A.R.); (P.R.); (C.F.); (D.H.); (P.R.)
- INSERM U1083, 49100 Angers, France
- Mitovasc Institute, Université d’Angers, 49100 Angers, France
| | - Daniel Henrion
- UMR CNRS 6015—INSERM U1083, IRIS2, 3 rue Roger Amsler, 49100 Angers, France; (J.M.C.d.l.B.); (A.R.); (P.R.); (C.F.); (D.H.); (P.R.)
- INSERM U1083, 49100 Angers, France
- Mitovasc Institute, Université d’Angers, 49100 Angers, France
- Angers University Hospital (CHU), 49100 Angers, France
| | - Pascal Reynier
- UMR CNRS 6015—INSERM U1083, IRIS2, 3 rue Roger Amsler, 49100 Angers, France; (J.M.C.d.l.B.); (A.R.); (P.R.); (C.F.); (D.H.); (P.R.)
- INSERM U1083, 49100 Angers, France
- Mitovasc Institute, Université d’Angers, 49100 Angers, France
- Service de Biochimie et Biologie Moléculaire, Centre Hospitalier Universitaire (CHU), 49000 Angers, France; (L.T.); (C.W.); (J.F.)
| | - Laurent Loufrani
- UMR CNRS 6015—INSERM U1083, IRIS2, 3 rue Roger Amsler, 49100 Angers, France; (J.M.C.d.l.B.); (A.R.); (P.R.); (C.F.); (D.H.); (P.R.)
- INSERM U1083, 49100 Angers, France
- Mitovasc Institute, Université d’Angers, 49100 Angers, France
- Correspondence: ; Tel.: +33-244688263
| |
Collapse
|
17
|
Targeting Arginine in COVID-19-Induced Immunopathology and Vasculopathy. Metabolites 2022; 12:metabo12030240. [PMID: 35323682 PMCID: PMC8953281 DOI: 10.3390/metabo12030240] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 01/27/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) represents a major public health crisis that has caused the death of nearly six million people worldwide. Emerging data have identified a deficiency of circulating arginine in patients with COVID-19. Arginine is a semi-essential amino acid that serves as key regulator of immune and vascular cell function. Arginine is metabolized by nitric oxide (NO) synthase to NO which plays a pivotal role in host defense and vascular health, whereas the catabolism of arginine by arginase to ornithine contributes to immune suppression and vascular disease. Notably, arginase activity is upregulated in COVID-19 patients in a disease-dependent fashion, favoring the production of ornithine and its metabolites from arginine over the synthesis of NO. This rewiring of arginine metabolism in COVID-19 promotes immune and endothelial cell dysfunction, vascular smooth muscle cell proliferation and migration, inflammation, vasoconstriction, thrombosis, and arterial thickening, fibrosis, and stiffening, which can lead to vascular occlusion, muti-organ failure, and death. Strategies that restore the plasma concentration of arginine, inhibit arginase activity, and/or enhance the bioavailability and potency of NO represent promising therapeutic approaches that may preserve immune function and prevent the development of severe vascular disease in patients with COVID-19.
Collapse
|
18
|
Single-nucleotide polymorphism of ADRβ2 and CDKN1B genes in Egyptian patients with coronary artery in-stent restenosis. Coron Artery Dis 2022; 33:277-283. [DOI: 10.1097/mca.0000000000001123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
19
|
Masi S, Ambrosini S, Mohammed SA, Sciarretta S, Lüscher TF, Paneni F, Costantino S. Epigenetic Remodeling in Obesity-Related Vascular Disease. Antioxid Redox Signal 2021; 34:1165-1199. [PMID: 32808539 DOI: 10.1089/ars.2020.8040] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The prevalence of obesity and cardiometabolic phenotypes is alarmingly increasing across the globe and is associated with atherosclerotic vascular complications and high mortality. In spite of multifactorial interventions, vascular residual risk remains high in this patient population, suggesting the need for breakthrough therapies. The mechanisms underpinning obesity-related vascular disease remain elusive and represent an intense area of investigation. Recent Advances: Epigenetic modifications-defined as environmentally induced chemical changes of DNA and histones that do not affect DNA sequence-are emerging as a potent modulator of gene transcription in the vasculature and might significantly contribute to the development of obesity-induced endothelial dysfunction. DNA methylation and histone post-translational modifications cooperate to build complex epigenetic signals, altering transcriptional networks that are implicated in redox homeostasis, mitochondrial function, vascular inflammation, and perivascular fat homeostasis in patients with cardiometabolic disturbances. Critical Issues: Deciphering the epigenetic landscape in the vasculature is extremely challenging due to the complexity of epigenetic signals and their function in regulating transcription. An overview of the most important epigenetic pathways is required to identify potential molecular targets to treat or prevent obesity-related endothelial dysfunction and atherosclerotic disease. This would enable the employment of precision medicine approaches in this setting. Future Directions: Current and future research efforts in this field entail a better definition of the vascular epigenome in obese patients as well as the unveiling of novel, cell-specific chromatin-modifying drugs that are able to erase specific epigenetic signals that are responsible for maladaptive transcriptional alterations and vascular dysfunction in obese patients. Antioxid. Redox Signal. 34, 1165-1199.
Collapse
Affiliation(s)
- Stefano Masi
- Dipartimento di Medicina Clinica e Sperimentale, Università di Pisa, Pisa, Italy
| | - Samuele Ambrosini
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Shafeeq A Mohammed
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| | - Sebastiano Sciarretta
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland.,Heart Division, Royal Brompton and Harefield Hospital Trust, National Heart & Lung Institute, Imperial College, London, United Kingdom
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Sarah Costantino
- Center for Molecular Cardiology, University of Zürich, Zurich, Switzerland
| |
Collapse
|
20
|
Douglass MS, Zhang Y, Kaplowitz MR, Fike CD. L-citrulline increases arginase II protein levels and arginase activity in hypoxic piglet pulmonary artery endothelial cells. Pulm Circ 2021; 11:20458940211006289. [PMID: 33948161 PMCID: PMC8053766 DOI: 10.1177/20458940211006289] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 03/08/2021] [Indexed: 11/15/2022] Open
Abstract
The L-arginine precursor, L-citrulline, re-couples endothelial nitric oxide synthase, increases nitric oxide production, and ameliorates chronic hypoxia-induced pulmonary hypertension in newborn pigs. L-arginine can induce arginase, which, in turn, may diminish nitric oxide production. Our major purpose was to determine if L-citrulline increases arginase activity in hypoxic piglet pulmonary arterial endothelial cells, and if so, concomitantly impacts the ability to increase endothelial nitric oxide synthase re-coupling and nitric oxide production. Piglet pulmonary arterial endothelial cells were cultured in hypoxic conditions with L-citrulline (0-3 mM) and/or the arginase inhibitor S-(2-boronoethyl)-L-cysteine. We measured arginase activity and nitric oxide production. We assessed endothelial nitric oxide synthase coupling by measuring endothelial nitric oxide synthase dimers and monomers. L-citrulline concentrations ≥0.5 mM increased arginase activity in hypoxic pulmonary arterial endothelial cells. L-citrulline concentrations ≥0.1 mM increased nitric oxide production and concentrations ≥0.5 mM elevated endothelial nitric oxide synthase dimer-to-monomer ratios. Co-treatment with L-citrulline and S-(2-boronoethyl)-L-cysteine elevated endothelial nitric oxide synthase dimer-to-monomer ratios more than sole treatment. Despite inducing arginase, L-citrulline increased nitric oxide production and endothelial nitric oxide synthase coupling in hypoxic piglet pulmonary arterial endothelial cells. However, these dose-dependent findings raise the possibility that there could be L-citrulline concentrations that elevate arginase to levels that negate improvements in endothelial nitric oxide synthase dysfunction. Moreover, our findings suggest that combining an arginase inhibitor with L-citrulline merits evaluation as a treatment for chronic hypoxia-induced pulmonary hypertension.
Collapse
Affiliation(s)
| | - Yongmei Zhang
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Mark R Kaplowitz
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Candice D Fike
- Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
21
|
Moretto J, Pudlo M, Demougeot C. Human-based evidence for the therapeutic potential of arginase inhibitors in cardiovascular diseases. Drug Discov Today 2020; 26:138-147. [PMID: 33197620 DOI: 10.1016/j.drudis.2020.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/22/2020] [Accepted: 11/05/2020] [Indexed: 01/25/2023]
Affiliation(s)
- Johnny Moretto
- PEPITE EA4267, FHU INCREASE, Université de Bourgogne Franche-Comté, F-25030 Besançon, France.
| | - Marc Pudlo
- PEPITE EA4267, FHU INCREASE, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| | - Céline Demougeot
- PEPITE EA4267, FHU INCREASE, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| |
Collapse
|
22
|
Liang Y, Alharthi AS, Elolimy AA, Bucktrout R, Lopreiato V, Martinez-Cortés I, Xu C, Fernandez C, Trevisi E, Loor JJ. Molecular networks of insulin signaling and amino acid metabolism in subcutaneous adipose tissue are altered by body condition in periparturient Holstein cows. J Dairy Sci 2020; 103:10459-10476. [PMID: 32921465 DOI: 10.3168/jds.2020-18612] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 07/05/2020] [Indexed: 12/28/2022]
Abstract
Peripartal cows mobilize not only body fat but also body protein to satisfy their energy requirements. The objective of this study was to determine the effect of prepartum BCS on blood biomarkers related to energy and nitrogen metabolism, and mRNA and protein abundance associated with AA metabolism and insulin signaling in subcutaneous adipose tissue (SAT) in peripartal cows. Twenty-two multiparous Holstein cows were retrospectively classified into a high BCS (HBCS; n = 11, BCS ≥ 3.5) or normal BCS (NBCS; n = 11, BCS ≤ 3.17) group at d 28 before expected parturition. Cows were fed the same diet as a total mixed ration before parturition and were fed the same lactation diet postpartum. Blood samples collected at -10, 7, 15, and 30 d relative to parturition were used for analyses of biomarkers associated with energy and nitrogen metabolism. Biopsies of SAT harvested at -15, 7, and 30 d relative to parturition were used for mRNA (real time-PCR) and protein abundance (Western blotting) assays. Data were subjected to ANOVA using the MIXED procedure of SAS (v. 9.4; SAS Institute Inc., Cary, NC), with P ≤ 0.05 being the threshold for significance. Cows in HBCS had greater overall plasma nonesterified fatty acid concentrations, due to marked increases at 7 and 15 d postpartum. This response was similar (BCS × Day effect) to protein abundance of phosphorylated (p) protein kinase B (p-AKT), the insulin-induced glucose transporter (SLC2A4), and the sodium-coupled neutral AA transporter (SLC38A1). Abundance of these proteins was lower at -15 d compared with NBCS cows, and either increased (SLC2A4, SLC38A1) or did not change (p-AKT) at 7 d postpartum in HBCS. Unlike protein abundance, however, overall mRNA abundances of the high-affinity cationic (SLC7A1), proton-coupled (SLC36A1), and sodium-coupled amino acid transporters (SLC38A2) were greater in HBCS than NBCS cows, due to upregulation in the postpartum phase. Those responses were similar to protein abundance of p-mTOR, which increased (BCS × Day effect) at 7 d in HBCS compared with NBCS cows. mRNA abundance of argininosuccinate lyase (ASL) and arginase 1 (ARG1) also was greater overall in HBCS cows. Together, these responses suggested impaired insulin signaling, coupled with greater postpartum AA transport rate and urea cycle activity in SAT of HBCS cows. An in vitro study using adipocyte and macrophage cocultures stimulated with various concentrations of fatty acids could provide some insights into the role of immune cells in modulating adipose tissue immunometabolic status, including insulin resistance and AA metabolism.
Collapse
Affiliation(s)
- Y Liang
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - A S Alharthi
- Department of Animal Production, College of Food and Agriculture Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - A A Elolimy
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock 72205; Arkansas Children's Nutrition Center, Little Rock 72205; Department of Animal Production, National Research Centre, Giza, 12611, Egypt
| | - R Bucktrout
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| | - V Lopreiato
- Department of Animal Sciences, Food and Nutrition, Faculty of Agriculture, Food and Environmental Science, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - I Martinez-Cortés
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801; Agricultural and Animal Production Department, UAM-Xochimilco, Mexico City 04960, Mexico
| | - C Xu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - C Fernandez
- Animal Science Department, Universitàt Politècnica de Valencia, 46022 Valencia, Spain
| | - E Trevisi
- Department of Animal Sciences, Food and Nutrition, Faculty of Agriculture, Food and Environmental Science, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - J J Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801.
| |
Collapse
|
23
|
Amino Acid Metabolism in Rheumatoid Arthritis: Friend or Foe? Biomolecules 2020; 10:biom10091280. [PMID: 32899743 PMCID: PMC7563518 DOI: 10.3390/biom10091280] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023] Open
Abstract
In mammals, amino acid metabolism has evolved to act as a critical regulator of innate and adaptive immune responses. Rheumatoid arthritis (RA) is the most common form of inflammatory arthropathy sustained by autoimmune responses. We examine here the current knowledge of tryptophan and arginine metabolisms and the main immunoregulatory pathways in amino acid catabolism, in both RA patients and experimental models of arthritis. We found that l-tryptophan (Trp) metabolism and, in particular, the kynurenine pathway would exert protective effects in all experimental models and in some, but not all, RA patients, possibly due to single nucleotide polymorphisms in the gene coding for indoleamine 2,3-dioxygenase 1 (IDO1; the enzyme catalyzing the rate-limiting step of the kynurenine pathway). The function, i.e., either protective or pathogenetic, of the l-arginine (Arg) metabolism in RA was less clear. In fact, although immunoregulatory arginase 1 (ARG1) was highly induced at the synovial level in RA patients, its true functional role is still unknown, possibly because of few available preclinical data. Therefore, our analysis would indicate that amino acid metabolism represents a fruitful area of research for new drug targets for a more effective and safe therapy of RA and that further studies are demanding to pursue such an important objective.
Collapse
|
24
|
S. Clemente G, van Waarde A, F. Antunes I, Dömling A, H. Elsinga P. Arginase as a Potential Biomarker of Disease Progression: A Molecular Imaging Perspective. Int J Mol Sci 2020; 21:E5291. [PMID: 32722521 PMCID: PMC7432485 DOI: 10.3390/ijms21155291] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/11/2022] Open
Abstract
Arginase is a widely known enzyme of the urea cycle that catalyzes the hydrolysis of L-arginine to L-ornithine and urea. The action of arginase goes beyond the boundaries of hepatic ureogenic function, being widespread through most tissues. Two arginase isoforms coexist, the type I (Arg1) predominantly expressed in the liver and the type II (Arg2) expressed throughout extrahepatic tissues. By producing L-ornithine while competing with nitric oxide synthase (NOS) for the same substrate (L-arginine), arginase can influence the endogenous levels of polyamines, proline, and NO•. Several pathophysiological processes may deregulate arginase/NOS balance, disturbing the homeostasis and functionality of the organism. Upregulated arginase expression is associated with several pathological processes that can range from cardiovascular, immune-mediated, and tumorigenic conditions to neurodegenerative disorders. Thus, arginase is a potential biomarker of disease progression and severity and has recently been the subject of research studies regarding the therapeutic efficacy of arginase inhibitors. This review gives a comprehensive overview of the pathophysiological role of arginase and the current state of development of arginase inhibitors, discussing the potential of arginase as a molecular imaging biomarker and stimulating the development of novel specific and high-affinity arginase imaging probes.
Collapse
Affiliation(s)
- Gonçalo S. Clemente
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| | - Inês F. Antunes
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| | - Alexander Dömling
- Department of Drug Design, Groningen Research Institute of Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands;
| | - Philip H. Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, The Netherlands; (G.S.C.); (A.v.W.); (I.F.A.)
| |
Collapse
|
25
|
Bujor A, Miron A, Luca SV, Skalicka-Wozniak K, Silion M, Trifan A, Girard C, Demougeot C, Totoson P. Vasorelaxant effects of Crataegus pentagyna: Links with arginase inhibition and phenolic profile. JOURNAL OF ETHNOPHARMACOLOGY 2020; 252:112559. [PMID: 31935497 DOI: 10.1016/j.jep.2020.112559] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 01/06/2020] [Accepted: 01/08/2020] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Crataegus leaves, flowers and fruits have been traditionally used to improve blood circulation, numerous preclinical and clinical studies supporting the cardiovascular benefits of Crataegus preparations. In this respect, there is very limited data on Crataegus pentagyna; in addition, the chemical profile of this species is still incompletely elucidated. AIM OF THE STUDY The objective of this study was to examine the cardiovascular benefits of Crataegus pentagyna Waldst. et Kit. ex Willd. (small-flowered black hawthorn, Rosaceae) extracts (leaf, flower and fruit ethyl acetate extracts) and the underlying mechanisms. We hypothesized that C. pentagyna extracts might exert vasodilatory effects and inhibit arginase activity due, in large part, to their polyphenolic constituents. MATERIALS AND METHODS C. pentagyna extracts induced-relaxation and the mechanisms involved were studied ex vivo in isolated aortic rings from Sprague-Dawley rats. The inhibitory effects on bovine liver arginase I were assessed by an in vitro assay. Metabolite profiling of C. pentagyna extracts was performed and the most endothelium- and nitric oxide synthase-dependent; flower extract additionally reduced Ca2+ entry and, to a lesser extent, Ca2+ release from the sarcoplasmic reticulum. C. pentagyna proved to be an important source of arginase inhibitors with potential benefits in endothelial dysfunction that remains to be explored.
Collapse
Affiliation(s)
- Alexandra Bujor
- Department of Pharmacognosy, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115, Iasi, Romania; PEPITE EA 4267, FHU INCREASE, University Bourgogne Franche-Comté, 19 rue Ambroise Paré, F-25030, Besançon, France.
| | - Anca Miron
- Department of Pharmacognosy, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115, Iasi, Romania.
| | - Simon Vlad Luca
- Department of Pharmacognosy, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115, Iasi, Romania; Biothermodynamics, TUM School of Life and Food Sciences Weihenstephan, Technical University of Munich, Maximus-von-Imhof-Forum 2, 85354, Freising, Germany.
| | - Krystyna Skalicka-Wozniak
- Department of Pharmacognosy with Medicinal Plant Unit, Medical University of Lublin, Chodzki 1, 20-093, Lublin, Poland.
| | - Mihaela Silion
- Petru Poni Institute of Macromolecular Chemistry, 41A Grigore Ghica Voda Alley, 700487, Iasi, Romania.
| | - Adriana Trifan
- Department of Pharmacognosy, Grigore T. Popa University of Medicine and Pharmacy, 16 Universitatii Street, 700115, Iasi, Romania.
| | - Corine Girard
- PEPITE EA 4267, FHU INCREASE, University Bourgogne Franche-Comté, 19 rue Ambroise Paré, F-25030, Besançon, France.
| | - Céline Demougeot
- PEPITE EA 4267, FHU INCREASE, University Bourgogne Franche-Comté, 19 rue Ambroise Paré, F-25030, Besançon, France.
| | - Perle Totoson
- PEPITE EA 4267, FHU INCREASE, University Bourgogne Franche-Comté, 19 rue Ambroise Paré, F-25030, Besançon, France.
| |
Collapse
|
26
|
Guerrouahen BS, Maccalli C, Cugno C, Rutella S, Akporiaye ET. Reverting Immune Suppression to Enhance Cancer Immunotherapy. Front Oncol 2020; 9:1554. [PMID: 32039024 PMCID: PMC6985581 DOI: 10.3389/fonc.2019.01554] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/23/2019] [Indexed: 01/26/2023] Open
Abstract
Tumors employ strategies to escape immune control. The principle aim of most cancer immunotherapies is to restore effective immune surveillance. Among the different processes regulating immune escape, tumor microenvironment-associated soluble factors, and/or cell surface-bound molecules are mostly responsible for dysfunctional activity of tumor-specific CD8+T cells. These dynamic immunosuppressive networks prevent tumor rejection at several levels, limiting also the success of immunotherapies. Nevertheless, the recent clinical development of immune checkpoint inhibitors or of molecules modulating cellular targets and immunosuppressive enzymes highlights the great potential of approaches based on the selective disruption of immunosuppressive networks. Currently, the administration of different categories of immunotherapy in combination regimens is the ultimate modality for impacting the survival of cancer patients. With the advent of immune checkpoint inhibitors, designed to mount an effective antitumor immune response, profound changes occurred in cancer immunotherapy: from a global stimulation of the immune system to a specific targeting of an immune component. This review will specifically highlight the players, the mechanisms limiting an efficient antitumor response and the current immunotherapy modalities tailored to target immune suppressive pathways. We also discuss the ongoing challenges encountered by these strategies and provide suggestions for circumventing hurdles to new immunotherapeutic approaches, including the use of relevant biomarkers in the optimization of immunotherapy regimens and the identification of patients who can benefit from defined immune-based approaches.
Collapse
Affiliation(s)
- Bella S Guerrouahen
- Sidra Medicine, Member of Qatar Foundation, Research Department, Doha, Qatar
| | - Cristina Maccalli
- Sidra Medicine, Member of Qatar Foundation, Research Department, Doha, Qatar
| | - Chiara Cugno
- Sidra Medicine, Member of Qatar Foundation, Research Department, Doha, Qatar
| | - Sergio Rutella
- John van Geest Cancer Research Centre, Nottingham Trent University, Nottingham, United Kingdom
| | - Emmanuel T Akporiaye
- Veana Therapeutics, Inc., Portland, OR, United States.,Providence Cancer Center, Portland, OR, United States
| |
Collapse
|
27
|
Durante W. Amino Acids in Circulatory Function and Health. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1265:39-56. [PMID: 32761569 DOI: 10.1007/978-3-030-45328-2_3] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease is the major cause of global mortality and disability. Abundant evidence indicates that amino acids play a fundamental role in cardiovascular physiology and pathology. Decades of research established the importance of L-arginine in promoting vascular health through the generation of the gas nitric oxide. More recently, L-glutamine, L-tryptophan, and L-cysteine have also been shown to modulate vascular function via the formation of a myriad of metabolites, including a number of gases (ammonia, carbon monoxide, hydrogen sulfide, and sulfur dioxide). These amino acids and their metabolites preserve vascular homeostasis by regulating critical cellular processes including proliferation, migration, differentiation, apoptosis, contractility, and senescence. Furthermore, they exert potent anti-inflammatory and antioxidant effects in the circulation, and block the accumulation of lipids within the arterial wall. They also mitigate known risk factors for cardiovascular disease, including hypertension, hyperlipidemia, obesity, and diabetes. However, in some instances, the metabolism of these amino acids through discrete pathways yields compounds that fosters vascular disease. While supplementation with amino acid monotherapy targeting the deficiency has ameliorated arterial disease in many animal models, this approach has been less successful in the clinic. A more robust approach combining amino acid supplementation with antioxidants, anti-inflammatory agents, and/or specific amino acid enzymatic pathway inhibitors may prove more successful. Alternatively, supplementation with amino acid-derived metabolites rather than the parent molecule may elicit beneficial effects while bypassing potentially harmful pathways of metabolism. Finally, there is an emerging recognition that circulating levels of multiple amino acids are perturbed in vascular disease and that a more holistic approach that targets all these amino acid derangements is required to restore circulatory function in diseased blood vessels.
Collapse
Affiliation(s)
- William Durante
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
28
|
Maternal Smoking Highly Affects the Function, Membrane Integrity, and Rheological Properties in Fetal Red Blood Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1509798. [PMID: 31871538 PMCID: PMC6906794 DOI: 10.1155/2019/1509798] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/25/2019] [Accepted: 10/15/2019] [Indexed: 01/13/2023]
Abstract
An understanding of the basic pathophysiological mechanisms of neonatal diseases necessitates detailed knowledge about the wide range of complications in the circulating fetal RBCs. Recent publications on adult red blood cells (RBCs) provide evidence that RBCs carry an active nitric oxide synthase (NOS3) enzyme and contribute to vascular functioning and integrity via their active nitric oxide synthesis. The aim of this study was to determine the effect of maternal smoking on the phenotypical appearance and functionality of fetal RBCs, based on morphological and molecular studies. We looked for possible links between vascular dysfunction and NOS3 expression and activation and its regulation by arginase (ARG1). Significant morphological and functional differences were found between fetal RBCs isolated from the arterial cord blood of neonates born to nonsmoking (RBC-NS, n = 62) and heavy-smoking (RBC-S, n = 51) mothers. Morphological variations were quantified by Advanced Cell Classifier, microscopy-based intelligent analysis software. To investigate the relevance of the newly suggested “erythrocrine” function in fetal RBCs, we measured the levels of NOS3 and its phosphorylation in parallel with the level of ARG1, as one of the major influencers of NOS3 dimerization, by fluorescence-activated cell sorting. Fetal RBCs, even the “healthy-looking” biconcave-shaped type, exhibited impaired NOS3 activation in the RBC-S population, which was paralleled with elevated ARG1 level, thus suggesting an increased redox burden. Our molecular data indicate that maternal smoking can exert marked effects on the circulating fetal RBCs, which could have a consequence on the outcome of in utero development. We hypothesize that any endothelial dysfunction altering NO production/bioavailability can be sensed by circulating fetal RBCs. Hence, we are putting forward the idea that neonatal RBC could serve as a real-time sensor for not only monitoring RBC-linked anomalies but also predicting the overall status of the vascular microenvironment.
Collapse
|
29
|
Mahdi A, Kövamees O, Pernow J. Improvement in endothelial function in cardiovascular disease - Is arginase the target? Int J Cardiol 2019; 301:207-214. [PMID: 31785959 DOI: 10.1016/j.ijcard.2019.11.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/26/2019] [Accepted: 11/04/2019] [Indexed: 01/30/2023]
Abstract
Endothelial dysfunction represents an early change in the vascular wall in areas prone to atherosclerotic plaque formation and is present in association with several risk factors for cardiovascular disease. The underlying mechanisms behind endothelial dysfunction are multifactorial and complex. Arginase has emerged as a key player in the regulation of endothelial integrity by the ability of reciprocally inhibits nitric oxide formation and promoting oxidative stress. A chain of evidence suggest that arginase is implicated in the pathogenesis underlying endothelial dysfunction induced by several cardiovascular risk factors and established cardiovascular disease including diabetes, hypercholesteremia, ischemia/reperfusion, atherosclerosis, obesity, ageing and hypertension. Recent data has unveiled a key role of arginase as one of the key mechanisms underlying endothelial dysfunction in diabetes and may serve as a potential therapeutic target in previously overlooked compartments including red blood cells. The current review is devoted to discuss arginase as a key mediator in endothelial dysfunction and the potential for therapeutic possibilities to target this enzyme in various diseases, especially type 2 diabetes, atherosclerosis and ischemia/reperfusion with focus on translational and clinical aspects. Moreover, approaches of how and in which patient group(s) arginase may be targeted in future clinical trials are discussed.
Collapse
Affiliation(s)
- Ali Mahdi
- Division of Cardiology, Department of Medicine, Division of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - Oskar Kövamees
- Division of Cardiology, Department of Medicine, Division of Cardiology, Karolinska Institutet, Stockholm, Sweden; Heart and Vascular Division, Karolinska University Hospital, Stockholm, Sweden
| | - John Pernow
- Division of Cardiology, Department of Medicine, Division of Cardiology, Karolinska Institutet, Stockholm, Sweden; Heart and Vascular Division, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
30
|
Metabolite profiling, arginase inhibition and vasorelaxant activity of Cornus mas, Sorbus aucuparia and Viburnum opulus fruit extracts. Food Chem Toxicol 2019; 133:110764. [PMID: 31437471 DOI: 10.1016/j.fct.2019.110764] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/08/2019] [Accepted: 08/15/2019] [Indexed: 12/18/2022]
Abstract
The present study investigated the effects of Cornus mas, Sorbus aucuparia and Viburnum opulus fruit extracts on arginase activity and arterial vasodilation. V. opulus fruit extract exerted the highest vasorelaxant activity in phenylephrine precontracted rat aortic rings (EC50 = 6.31 ± 1.61 μg/mL) and a significant inhibition of arginase (IC50 = 71.02 ± 3.06 μg/mL). By contrast, S. aucuparia and C. mas fruit extracts showed no important anti-arginase activity and a significantly weaker activity in the rat aortic rings relaxation assay (EC50 = 100.9 ± 11.63 and 78.52 ± 8.59 μg/mL, respectively). For all extracts, the main mechanism of vasodilation was proven to be endothelium-dependent. HPLC-ESI-Q-TOF-MS/MS studies revealed a very complex metabolite profiling in all three extracts with chlorogenic acid accounting for 30.89, 0.72 and 2.03 mg/g in V. opulus, C. mas and S. aucuparia fruit extracts, respectively. All extracts were declared non-toxic in the brine shrimp acute toxicity test. Our study highlights potential benefits of V. opulus fruit extract in diseases associated with endothelial dysfunction and impaired vasodilation.
Collapse
|
31
|
Seo C, Kim SH, Lee HS, Ji M, Min J, Son YJ, Kim IH, Lee K, Paik MJ. Metabolomic study on bleomycin and polyhexamethylene guanidine phosphate-induced pulmonary fibrosis mice models. Metabolomics 2019; 15:111. [PMID: 31422500 DOI: 10.1007/s11306-019-1574-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 08/01/2019] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Polyhexamethylene guanidine phosphate (PHMG) has been used as a disinfectant and biocide, and was known to be harmless and non-toxic. However, in 2011, PHMG used as a humidifier disinfectant was reported to be associated with lung diseases, such as, fibrosis in the toxicant studies on pulmonary fibrosis by PHMG. However, no metabolomics study has been performed in PHMG-induced mouse models of pulmonary fibrosis. OBJECTIVES We performed a metabolomic study to understand the biochemical events that occur in bleomycin (BLM)- and PHMG-induced mouse models of pulmonary fibrosis using gas chromatography-mass spectrometry (GC-MS), LC-tandem MS, and GC-tandem MS. RESULTS The levels of 61 metabolites of 30 amino acids, 13 organic acids, 12 fatty acids, 5 polyamines, and oxidized glutathione were determined in the pulmonary tissues of mice with BLM- and PHMG-induced pulmonary fibrosis and in normal controls. Principal component analysis and partial least squares discriminant analysis used to compare level of these 61 metabolites in pulmonary tissues. Levels of metabolites were significantly different in the BLM and PHMG groups as compared with the control group. In particular, the BLM- and PHMG-induced pulmonary fibrosis models showed elevated collagen synthesis and oxidative stress and metabolic disturbance of TCA related organic acids including fumaric acid by NADPH oxidase. In addition, polyamine metabolism showed severe alteration in the PHMG group than that of the BLM group. CONCLUSION This result suggests PHMG will be able to induce pulmonary fibrosis by arginine metabolism and NADPH oxidase signaling.
Collapse
Affiliation(s)
- Chan Seo
- College of Pharmacy, Sunchon National University, Suncheon, 540-950, Republic of Korea
| | - Sung-Hwan Kim
- National Center for Efficacy Evaluation of Respiratory Disease Product, Institute of Toxicology, Jeongeup-si, 56212, Republic of Korea
| | - Hyeon-Seong Lee
- College of Pharmacy, Sunchon National University, Suncheon, 540-950, Republic of Korea
| | - Moongi Ji
- College of Pharmacy, Sunchon National University, Suncheon, 540-950, Republic of Korea
| | - Jeuk Min
- College of Pharmacy, Sunchon National University, Suncheon, 540-950, Republic of Korea
| | - Young-Jin Son
- College of Pharmacy, Sunchon National University, Suncheon, 540-950, Republic of Korea
| | - In-Hyeon Kim
- National Center for Efficacy Evaluation of Respiratory Disease Product, Institute of Toxicology, Jeongeup-si, 56212, Republic of Korea
| | - Kyuhong Lee
- National Center for Efficacy Evaluation of Respiratory Disease Product, Institute of Toxicology, Jeongeup-si, 56212, Republic of Korea.
| | - Man-Jeong Paik
- College of Pharmacy, Sunchon National University, Suncheon, 540-950, Republic of Korea.
| |
Collapse
|
32
|
Chandrasekharan UM, Wang Z, Wu Y, Wilson Tang WH, Hazen SL, Wang S, Elaine Husni M. Elevated levels of plasma symmetric dimethylarginine and increased arginase activity as potential indicators of cardiovascular comorbidity in rheumatoid arthritis. Arthritis Res Ther 2018; 20:123. [PMID: 29884228 PMCID: PMC5994036 DOI: 10.1186/s13075-018-1616-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 05/08/2018] [Indexed: 12/20/2022] Open
Abstract
Background Rheumatoid arthritis (RA) patients are at high risk of developing cardiovascular disease (CVD). In RA, chronic inflammation may lead to endothelial dysfunction, an early indicator of CVD, owing to diminished nitric oxide (NO) production. Because l-arginine is the sole precursor of NO, we hypothesized that levels of l-arginine metabolic products reflecting NO metabolism are altered in patients with RA. Methods Plasma samples from patients with RA (n = 119) and age- and sex-matched control subjects (n = 238) were used for this study. Using LC-MS/MS, we measured plasma levels of free l-arginine, l-ornithine, l-citrulline, l-NG-monomethyl arginine (MMA), asymmetric dimethylarginine (ADMA), and symmetric dimethylarginine (SDMA). We compared global arginine bioavailability ratio (GABR) (i.e., ratio of l-arginine to l-ornithine + l-citrulline) and arginine methylation index (ArgMI) (i.e., ADMA + SDMA/MMA) in patients with RA vs. control subjects. Plasma arginase activity was measured using a sensitive arginase assay kit. The relationship of l-arginine metabolites and arginase activity to CVD risk factors was evaluated using Pearson’s chi-square test. Results Compared with healthy control subjects, the RA cohort showed significantly lower levels of plasma l-arginine (46.11 ± 17.29 vs. 74.2 ± 22.53 μmol/L, p < 0.001) and GABR (0.36 ± 0.16 vs. 0.73 ± 0.24, p < 0.001), elevated levels of ADMA (0.76 ± 0.12 vs. 0.62 ± 0.12 μmol/L, p < 0.001), SDMA (0.54 ± 0.14 vs. 0.47 ± 0.13 μmol/L, p < 0.001), and ArgMI (6.51 ± 1.86 vs. 5.54 ± 1.51, p < 0.001). We found an approximately fourfold increase in arginase activity (33.8 ± 1.1 vs. 8.4 ± 0.8 U/L, p < 0.001), as well as elevated levels of arginase-mediated l-arginine catalytic product l-ornithine (108.64 ± 30.26 vs. 69.3 ± 20.71 μmol/L, p < 0.001), whereas a nitric oxide synthase (NOS) catalytic product, the l-citrulline level, was diminished in RA (30.32 ± 9.93 vs. 36.17 ± 11.64 μmol/L, p < 0.001). Patients with RA with existing CVD had higher arginase activity than patients with RA without CVD (p = 0.048). Conclusions Global l-arginine bioavailability was diminished, whereas plasma arginase activity, ADMA, and SDMA levels were elevated, in patients with RA compared with healthy control subjects. Plasma SDMA was associated with hypertension and hyperlipidemia in patients with RA. This dysregulated l-arginine metabolism may function as a potential indicator of CVD risk in patients with RA.
Collapse
Affiliation(s)
| | - Zeneng Wang
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Yuping Wu
- Department of Mathematics, Cleveland State University, Cleveland, OH, USA
| | - W H Wilson Tang
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Cardiovascular Disease, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Stanley L Hazen
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Cardiovascular Disease, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Sihe Wang
- Department of Laboratory Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - M Elaine Husni
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA. .,Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
33
|
Cooke EJ, Zhou JY, Wyseure T, Joshi S, Bhat V, Durden DL, Mosnier LO, von Drygalski A. Vascular Permeability and Remodelling Coincide with Inflammatory and Reparative Processes after Joint Bleeding in Factor VIII-Deficient Mice. Thromb Haemost 2018; 118:1036-1047. [PMID: 29847841 PMCID: PMC6191040 DOI: 10.1055/s-0038-1641755] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Vascular remodelling is a prominent feature of haemophilic arthropathy (HA) that may underlie re-bleeding, yet the nature of vascular changes and underlying mechanisms remain largely unknown. Here, we aimed to characterize synovial vascular remodelling and vessel integrity after haemarthrosis, as well as temporal changes in inflammatory and tissue-reparative pathways. Thirty acutely painful joints in patients with haemophilia (PWH) were imaged by musculoskeletal ultrasound with Power Doppler (MSKUS/PD) to detect vascular abnormalities and bloody effusions. Nineteen out of 30 painful joint episodes in PWH were associated with haemarthrosis, and abnormal vascular perfusion was unique to bleeding joints. A model of induced haemarthrosis in factor VIII (FVIII)-deficient mice was used for histological assessment of vascular remodelling (α-smooth muscle actin [αSMA] expression), and monitoring of in vivo vascular perfusion and permeability by MSKUS/PD and albumin extravasation, respectively. Inflammatory (M1) and reparative (M2) macrophage markers were quantified in murine synovium over a 10-week time course by real-time polymerase chain reaction. The abnormal vascular perfusion observed in PWH was recapitulated in FVIII-deficient mice after induced haemarthrosis. Neovascularization and increased vessel permeability were apparent 2 weeks post-bleed in FVIII-deficient mice, after a transient elevation of inflammatory macrophage M1 markers. These vascular changes subsided by week 4, while vascular remodelling, evidenced by architectural changes and pronounced αSMA expression, persisted alongside a reparative macrophage M2 response. In conclusion, haemarthrosis leads to transient inflammation coupled with neovascularization and associated vascular permeability, while subsequent tissue repair mechanisms coincide with vascular remodelling. Together, these vascular changes may promote re-bleeding and HA progression.
Collapse
Affiliation(s)
- Esther J Cooke
- University of California San Diego, Department of Medicine, Division of Hematology/Oncology, La Jolla, CA, USA
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Jenny Y Zhou
- University of California San Diego, Department of Medicine, Division of Hematology/Oncology, La Jolla, CA, USA
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Tine Wyseure
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Shweta Joshi
- University of California San Diego, Department of Pediatrics, Moores UCSD Cancer Center, La Jolla, CA, USA
| | - Vikas Bhat
- University of California San Diego, Department of Medicine, Division of Hematology/Oncology, La Jolla, CA, USA
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Donald L Durden
- University of California San Diego, Department of Pediatrics, Moores UCSD Cancer Center, La Jolla, CA, USA
| | - Laurent O Mosnier
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| | - Annette von Drygalski
- University of California San Diego, Department of Medicine, Division of Hematology/Oncology, La Jolla, CA, USA
- The Scripps Research Institute, Department of Molecular Medicine, La Jolla, CA, USA
| |
Collapse
|
34
|
López V, Moraga FA, Llanos AJ, Ebensperger G, Taborda MI, Uribe E. Plasmatic Concentrations of ADMA and Homocystein in Llama ( Lama glama) and Regulation of Arginase Type II: An Animal Resistent to the Development of Pulmonary Hypertension Induced by Hypoxia. Front Physiol 2018; 9:606. [PMID: 29896110 PMCID: PMC5986928 DOI: 10.3389/fphys.2018.00606] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 05/04/2018] [Indexed: 12/21/2022] Open
Abstract
There are animal species that have adapted to life at high altitude and hypobaric hypoxia conditions in the Andean highlands. One such species is the llama (Lama glama), which seem to have developed efficient protective mechanisms to avoid maladaptation resulting from chronic hypoxia, such as a resistance to the development of hypoxia -induced pulmonary hypertension. On the other hand, it is widely known that different models of hypertension can arise as a result of changes in endothelial function. The respect, one of the common causes of deregulation in endothelial vasodilator function have been associated with down-regulation of the NO synthesis and an increase in plasma levels of asymmetric dimethylarginine (ADMA) and homocysteine. Additionally, it is also known that NO production can be regulated by plasma levels of L-arginine as a result of the competition between nitric oxide synthase (NOS) and arginase. The objective of this study, was to determine the baseline concentrations of ADMA and homocysteine in llama, and to evaluate their effect on the arginase pathway and their involvement in the resistance to the development of altitude-induced pulmonary hypertension. METHOD Lowland and highland newborn sheep and llama were investigated near sea level and at high altitude. Blood determinations of arterial blood gases, ADMA and homocysteíne are made and the effect of these on the arginase activity was evaluated. RESULTS The basal concentrations of ADMA and homocysteine were determined in llama, and they were found to be significantly lower than those found in other species and in addition, the exposure to hypoxia is unable to increase its concentration. On the other hand, it was observed that the llama exhibited 10 times less arginase II activity as compared to sheep, and the expression was not induced by hypoxia. Finally, ADMA y Hcy, has no effect on the type II arginase pathway. CONCLUSION Based on our results, we propose that low concentrations of ADMA and homocysteine found in llamas, the low expression of arginase type II, DDAH-2 and CBS, as well as its insensitivity to activation by homocysteine could constitute an adaptation mechanism of these animals to the hypoxia.
Collapse
Affiliation(s)
- Vasthi López
- Laboratorio de Metabolismo de Aminoácidos e Hipoxia, Departamento de Ciencias Biomédicas, Universidad Católica del Norte, Coquimbo, Chile
| | - Fernando A Moraga
- Laboratorio de Fisiología, Hipoxia y Función Vascular, Departamento de Ciencias Biomedicas, Facultad de Medicina, Universidad Católica del Norte, Coquimbo, Chile
| | - Anibal J Llanos
- Laboratorio de Fisiología y Fisiopatología del Desarrollo, Departamento de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - German Ebensperger
- Laboratorio de Fisiología y Fisiopatología del Desarrollo, Departamento de Ciencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - María I Taborda
- Laboratorio de Metabolismo de Aminoácidos e Hipoxia, Departamento de Ciencias Biomédicas, Universidad Católica del Norte, Coquimbo, Chile
| | - Elena Uribe
- Laboratorio de Enzimología, Departamento de Bioquímica y Biología Molecular, Universidad of Concepción, Concepción, Chile
| |
Collapse
|
35
|
Peyton KJ, Liu XM, Shebib AR, Johnson FK, Johnson RA, Durante W. Arginase inhibition prevents the development of hypertension and improves insulin resistance in obese rats. Amino Acids 2018; 50:747-754. [PMID: 29700652 DOI: 10.1007/s00726-018-2567-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 04/09/2018] [Indexed: 01/12/2023]
Abstract
This study investigated the temporal activation of arginase in obese Zucker rats (ZR) and determined if arginase inhibition prevents the development of hypertension and improves insulin resistance in these animals. Arginase activity, plasma arginine and nitric oxide (NO) concentration, blood pressure, and insulin resistance were measured in lean and obese animals. There was a chronological increase in vascular and plasma arginase activity in obese ZR beginning at 8 weeks of age. The increase in arginase activity in obese animals was associated with a decrease in insulin sensitivity and circulating levels of arginine and NO. The rise in arginase activity also preceded the increase in blood pressure in obese ZR detected at 12 weeks of age. Chronic treatment of 8-week-old obese animals with an arginase inhibitor or L-arginine for 4 weeks prevented the development of hypertension and improved plasma concentrations of arginine and NO. Arginase inhibition also improved insulin sensitivity in obese ZR while L-arginine supplementation had no effect. In conclusion, arginase inhibition prevents the development of hypertension and improves insulin sensitivity while L-arginine administration only mitigates hypertension in obese animals. Arginase represents a promising therapeutic target in ameliorating obesity-associated vascular and metabolic dysfunction.
Collapse
Affiliation(s)
- Kelly J Peyton
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, M409 Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Xiao-Ming Liu
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, M409 Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Ahmad R Shebib
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, M409 Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA
| | - Fruzsina K Johnson
- College of Osteopathic Medicine, William Cary University, Hattiesburg, MS, USA
| | - Robert A Johnson
- College of Osteopathic Medicine, William Cary University, Hattiesburg, MS, USA
| | - William Durante
- Department of Medical Pharmacology and Physiology, School of Medicine, University of Missouri, M409 Medical Sciences Building, One Hospital Drive, Columbia, MO, 65212, USA.
| |
Collapse
|
36
|
Caldwell RW, Rodriguez PC, Toque HA, Narayanan SP, Caldwell RB. Arginase: A Multifaceted Enzyme Important in Health and Disease. Physiol Rev 2018; 98:641-665. [PMID: 29412048 PMCID: PMC5966718 DOI: 10.1152/physrev.00037.2016] [Citation(s) in RCA: 290] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 08/14/2017] [Accepted: 08/17/2017] [Indexed: 12/15/2022] Open
Abstract
The arginase enzyme developed in early life forms and was maintained during evolution. As the last step in the urea cycle, arginase cleaves l-arginine to form urea and l-ornithine. The urea cycle provides protection against excess ammonia, while l-ornithine is needed for cell proliferation, collagen formation, and other physiological functions. In mammals, increases in arginase activity have been linked to dysfunction and pathologies of the cardiovascular system, kidney, and central nervous system and also to dysfunction of the immune system and cancer. Two important aspects of the excessive activity of arginase may be involved in diseases. First, overly active arginase can reduce the supply of l-arginine needed for the production of nitric oxide (NO) by NO synthase. Second, too much l-ornithine can lead to structural problems in the vasculature, neuronal toxicity, and abnormal growth of tumor cells. Seminal studies have demonstrated that increased formation of reactive oxygen species and key inflammatory mediators promote this pathological elevation of arginase activity. Here, we review the involvement of arginase in diseases affecting the cardiovascular, renal, and central nervous system and cancer and discuss the value of therapies targeting the elevated activity of arginase.
Collapse
Affiliation(s)
- R William Caldwell
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - Paulo C Rodriguez
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - Haroldo A Toque
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - S Priya Narayanan
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - Ruth B Caldwell
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| |
Collapse
|
37
|
Abstract
Over the past century, solid organ transplantation has been improved both at a surgical and postoperative level. However, despite the improvement in efficiency, safety, and survival, we are still far from obtaining full acceptance of all kinds of allograft in the absence of concomitant treatments. Today, transplanted patients are treated with immunosuppressive drugs (IS) to minimize immunological response in order to prevent graft rejection. Nevertheless, the lack of specificity of IS leads to an increase in the risk of cancer and infections. At this point, cell therapies have been shown as a novel promising resource to minimize the use of IS in transplantation. The main strength of cell therapy is the opportunity to generate allograft-specific tolerance, promoting in this way long-term allograft survival. Among several other regulatory cell types, tolerogenic monocyte-derived dendritic cells (Tol-MoDCs) appear to be an interesting candidate for cell therapy due to their ability to perform specific antigen presentation and to polarize immune response to immunotolerance. In this review, we describe the characteristics and the mechanisms of action of both human Tol-MoDCs and rodent tolerogenic bone marrow-derived DCs (Tol-BMDCs). Furthermore, studies performed in transplantation models in rodents and non-human primates corroborate the potential of Tol-BMDCs for immunoregulation. In consequence, Tol-MoDCs have been recently evaluated in sundry clinical trials in autoimmune diseases and shown to be safe. In addition to autoimmune diseases clinical trials, Tol-MoDC is currently used in the first phase I/II clinical trials in transplantation. Translation of Tol-MoDCs to clinical application in transplantation will also be discussed in this review.
Collapse
Affiliation(s)
- Eros Marín
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Nephrologie (ITUN), CHU Nantes, Nantes, France
| | - Maria Cristina Cuturi
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Nephrologie (ITUN), CHU Nantes, Nantes, France
| | - Aurélie Moreau
- Centre de Recherche en Transplantation et Immunologie UMR1064, INSERM, Université de Nantes, Nantes, France.,Institut de Transplantation Urologie Nephrologie (ITUN), CHU Nantes, Nantes, France
| |
Collapse
|
38
|
Chen M, Zhao J, Ali IHA, Marry S, Augustine J, Bhuckory M, Lynch A, Kissenpfennig A, Xu H. Cytokine Signaling Protein 3 Deficiency in Myeloid Cells Promotes Retinal Degeneration and Angiogenesis through Arginase-1 Up-Regulation in Experimental Autoimmune Uveoretinitis. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:1007-1020. [PMID: 29452101 DOI: 10.1016/j.ajpath.2017.12.021] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 11/14/2017] [Accepted: 12/07/2017] [Indexed: 01/01/2023]
Abstract
The suppressor of cytokine signaling protein 3 (SOCS3) critically controls immune cell activation, although its role in macrophage polarization and function remains controversial. Using experimental autoimmune uveoretinitis (EAU) as a model, we show that inflammation-mediated retinal degeneration is exaggerated and retinal angiogenesis is accelerated in mice with SOCS3 deficiency in myeloid cells (LysMCre/+SOCS3fl/fl). At the acute stage of EAU, the population of infiltrating neutrophils was increased and the population of macrophages decreased in LysMCre/+SOCS3fl/fl mice compared with that in wild-type (WT) mice. Real-time RT-PCR showed that the expression of tumor necrosis factor-α, IL-1β, interferon-γ, granulocyte-macrophage colony-stimulating factor, and arginase-1 was significantly higher in the LysMCre/+SOCS3fl/fl EAU retina in contrast to the WT EAU retina. The percentage of arginase-1+ infiltrating cells was significantly higher in the LysMCre/+SOCS3fl/fl EAU retina than that in the WT EAU retina. In addition, bone marrow-derived macrophages and neutrophils from the LysMCre/+SOCS3fl/fl mice express significantly higher levels of chemokine (C-C motif) ligand 2 and arginase-1 compared with those from WT mice. Inhibition of arginase using an l-arginine analog amino-2-borono-6-hexanoic suppressed inflammation-induced retinal angiogenesis without affecting the severity of inflammation. Our results suggest that SOCS3 critically controls the phenotype and function of macrophages and neutrophils under inflammatory conditions and loss of SOCS3 promotes the angiogenic phenotype of the cells through up-regulation of arginase-1.
Collapse
Affiliation(s)
- Mei Chen
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Jiawu Zhao
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Imran H A Ali
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Stephen Marry
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Josy Augustine
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Mohajeet Bhuckory
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Aisling Lynch
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Adrien Kissenpfennig
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom
| | - Heping Xu
- Centre for Experimental Medicine, School of Medicine, Dentistry & Biomedical Science, Queen's University Belfast, Belfast, United Kingdom.
| |
Collapse
|
39
|
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
|
40
|
Polyamine concentration is increased in thoracic ascending aorta of patients with bicuspid aortic valve. Heart Vessels 2017; 33:327-339. [DOI: 10.1007/s00380-017-1087-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/13/2017] [Indexed: 12/26/2022]
|
41
|
Souza ÁDPBD, Oliveira MMRD, Andrade RRD, Amorim RFBD, Bocca AL, Borin MDF. The in vivo effect of L-arginine on skin elasticity in mice. BRAZ J PHARM SCI 2017. [DOI: 10.1590/s2175-97902017000300045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
42
|
Lee SJ, Lee JS, Choi MH, Lee SE, Shin DH, Hong JM. Cilostazol improves endothelial function in acute cerebral ischemia patients: a double-blind placebo controlled trial with flow-mediated dilation technique. BMC Neurol 2017; 17:169. [PMID: 28851320 PMCID: PMC5576326 DOI: 10.1186/s12883-017-0950-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 08/24/2017] [Indexed: 02/07/2023] Open
Abstract
Background In order to evaluate the impact of cilostazol on endothelial function, we compared the changes of flow-mediated dilation (FMD) between aspirin and cilostazol groups in patients with acute cerebral ischemia. Methods Patients presenting with acute cerebral ischemic events were randomly assigned into aspirin (n = 40) or cilostazol (n = 40) group in a double-blinded manner. FMD was measured at baseline (T0) and 90 days (T1). We measured L-arginine at baseline (a precursor of biologically active nitric oxides). Serious and non-serious adverse events were described. Results Despite no difference in the baseline FMD values (p = 0.363), there was a significant increase of FMD values in cilostazol group (7.9 ± 2.4 to 8.9 ± 2.3%, p = 0.001) and not in aspirin group (8.5 ± 2.6 to 9.3 ± 2.8%, p = 0.108). In the multiple regression analysis performed in cilostazol group, serum L–arginine levels were inversely correlated with FMD at T1 (ß = −0.050, SE: 0.012, p < 0.001) with age, total cholesterol levels, and C-reactive protein as confounders. While T0 FMD values in both aspirin and cilostazol groups did not show any correlation with serum L-arginine levels, the correlation is restored in the cilostazol group at T1 (r = 0.467, p = 0.007), while such is not shown in the aspirin group. There was no difference of serious adverse events between the two groups (p = 0.235). Adverse events were more common in the cilostazol group (35/40 vs. 25/40, p = 0.010), due to frequent headaches (14/40 vs. 3/30, p = 0.003) which was well tolerated. Conclusion Cilostazol improved endothelial function in acute cerebral ischemia patients. It also restored an inverse correlation between 3-month FMD and baseline L-arginine levels. Trial registration NCT03116269, 04/12/2017, retrospectively registered. Electronic supplementary material The online version of this article (10.1186/s12883-017-0950-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Seong-Joon Lee
- Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Jin Soo Lee
- Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Mun Hee Choi
- Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Sung Eun Lee
- Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Dong Hoon Shin
- Department of Neurology, Gachon University Gil Hospital, Incheon, Republic of Korea
| | - Ji Man Hong
- Department of Neurology, Ajou University School of Medicine, Suwon, Republic of Korea. .,Department of Neurology, Ajou University School of Medicine, 164, World cup-ro, Yeongtong-gu, Suwon-si, Gyeonggi-do, 16499, South Korea.
| |
Collapse
|
43
|
Arginase Inhibition Reverses Monocrotaline-Induced Pulmonary Hypertension. Int J Mol Sci 2017; 18:ijms18081609. [PMID: 28757567 PMCID: PMC5578001 DOI: 10.3390/ijms18081609] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 07/13/2017] [Accepted: 07/20/2017] [Indexed: 01/19/2023] Open
Abstract
Pulmonary hypertension (PH) is a heterogeneous disorder associated with a poor prognosis. Thus, the development of novel treatment strategies is of great interest. The enzyme arginase (Arg) is emerging as important player in PH development. The aim of the current study was to determine the expression of ArgI and ArgII as well as the effects of Arg inhibition in a rat model of PH. PH was induced in 35 Sprague–Dawley rats by monocrotaline (MCT, 60 mg/kg as single-dose). There were three experimental groups: sham-treated controls (control group, n = 11), MCT-induced PH (MCT group, n = 11) and MCT-induced PH treated with the Arg inhibitor Nω-hydroxy-nor-l-arginine (nor-NOHA; MCT/NorNoha group, n = 13). ArgI and ArgII expression was determined by immunohistochemistry and Western blot. Right ventricular systolic pressure (RVPsys) was measured and lung tissue remodeling was determined. Induction of PH resulted in an increase in RVPsys (81 ± 16 mmHg) compared to the control group (41 ± 15 mmHg, p = 0.002) accompanied by a significant elevation of histological sum-score (8.2 ± 2.4 in the MCT compared to 1.6 ± 1.6 in the control group, p < 0.001). Both, ArgI and ArgII were relevantly expressed in lung tissue and there was a significant increase in the MCT compared to the control group (p < 0.01). Arg inhibition resulted in a significant reduction of RVPsys to 52 ± 19 mmHg (p = 0.006) and histological sum-score to 5.8 ± 1.4 compared to the MCT group (p = 0.022). PH leads to increased expression of Arg. Arg inhibition leads to reduction of RVPsys and diminished lung tissue remodeling and therefore represents a potential treatment strategy in PH.
Collapse
|
44
|
Plasma l-citrulline concentrations in l-arginine-supplemented healthy dogs. J Vet Cardiol 2017; 19:376-383. [PMID: 28684243 DOI: 10.1016/j.jvc.2017.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 04/11/2017] [Accepted: 04/18/2017] [Indexed: 11/23/2022]
Abstract
INTRODUCTION To determine whether oral l-arginine increases plasma [l-citrulline] in dogs. ANIMALS Eleven healthy staff-owned dogs were used in this study. MATERIALS AND METHODS Dogs (n = 3) were given l-arginine (50mg/kg PO q8h) for 7 days, and plasma [l-arginine] and [l-citrulline] were analyzed by high performance liquid chromatography at baseline (BL), steady state trough, and 0.5, 1, 1.5, 2, 4, 6, and 8 h after final dosing on day 7. Eleven dogs were then treated with 100mg/kg l-arginine PO q8h for 7 days, and [l-arginine] and [l-citrulline] were measured at BL, steady state trough, and at peak 4 hrs after dosing (T4 hrs). RESULTS - Plasma [l-arginine] and [l-citrulline] peaked at T4 hrs on the 50mg/kg dosage. Target outcome, modeled after human study results, of a doubling of [l-arginine] and a 25-30% increase in [l-citrulline] from BL were not reached. After the 100mg/kg dosage, plasma [l-arginine] increased from a BL median of 160.1 μM (range, 100.2-231.4 μM) to a peak of 417.4 μM (206.5-807.3 μM) at T4 hrs, and plasma [l-citrulline] increased from a BL median of 87.8 μM (59.1-117.1 μM) to peak of 102.2 μM (47.4-192.6 μM) at T4 hrs. Ten of eleven dogs showed a doubling of plasma [l-arginine] and 4/11 dogs achieved 25-30% or greater increases in plasma [l-citrulline]. No adverse effects on heart rate or blood pressure were noted. CONCLUSIONS - Oral l-arginine dosage of 100mg/kg q8h doubles plasma [l-arginine] in healthy dogs, but conversion to l-citrulline is quite variable. Further evaluation of this dosage regimen in dogs with pulmonary hypertension is warranted.
Collapse
|
45
|
Pudlo M, Demougeot C, Girard-Thernier C. Arginase Inhibitors: A Rational Approach Over One Century. Med Res Rev 2016; 37:475-513. [DOI: 10.1002/med.21419] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/14/2016] [Accepted: 09/22/2016] [Indexed: 12/28/2022]
Affiliation(s)
- Marc Pudlo
- PEPITE - EA4267; University Bourgogne Franche-Comté; Besançon France
| | - Céline Demougeot
- PEPITE - EA4267; University Bourgogne Franche-Comté; Besançon France
| | | |
Collapse
|
46
|
Arginase inhibition attenuates arteriogenesis and interferes with M2 macrophage accumulation. J Transl Med 2016; 96:830-8. [PMID: 27239731 DOI: 10.1038/labinvest.2016.62] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 04/05/2016] [Accepted: 04/21/2016] [Indexed: 01/27/2023] Open
Abstract
l-Arginine is the common substrate for nitric oxide synthases (NOS) and arginase. Whereas the contribution of NOS to collateral artery growth (arteriogenesis) has been demonstrated, the functional role of arginase remains to be elucidated and was topic of the present study. Arteriogenesis was induced in mice by ligation of the femoral artery. Laser Doppler perfusion measurements demonstrated a significant reduction in arteriogenesis in mice treated with the arginase inhibitor nor-NOHA (N(ω)-hydroxy-nor-arginine). Accompanying in vitro results on murine primary arterial endothelial cells and smooth muscle cells revealed that nor-NOHA treatment interfered with cell proliferation and resulted in increased nitrate/nitrite levels, indicative for increased NO production. Immuno-histological analyses on tissue samples demonstrated that nor-NOHA administration caused a significant reduction in M2 macrophage accumulation around growing collateral arteries. Gene expression studies on isolated growing collaterals evidenced that nor-NOHA treatment abolished the differential expression of Icam1 (intercellular adhesion molecule 1). From our data we conclude that arginase activity is essential for arteriogenesis by promoting perivascular M2 macrophage accumulation as well as arterial cell proliferation.
Collapse
|
47
|
Ciuntu A. Role of Metabolites of Nitric Oxide and Arginase in the Pathogenesis of Glomerulonephritis. CURRENT HEALTH SCIENCES JOURNAL 2016; 42:221-225. [PMID: 30581575 PMCID: PMC6269603 DOI: 10.12865/chsj.42.03.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/01/2016] [Indexed: 11/18/2022]
Abstract
PURPOSE The aim of the study is to assess the level of nitric oxide metabolites and arginase in the urine of children with glomerulonephritis depending on clinical evolutional stages of the disease. MATERIALS AND METHODS The prospective study included 65 children with primary glomerulonephritis, 25 children with steroid-sensitive nephrotic syndrome (SSNS) and 20 children with steroid-resistant nephrotic syndrome (SRNS), 20 children with mixed form of chronic glomerulonephritis(CGN). RESULTS Thus in the SRNS group, during relapse period the concentration of NO metabolites in urine was increased by 4,2 times, while in SSNS by 3,0 times in comparison with the control group. The concentration of NO metabolites in the urine increased by 4,8 times during relapse CGN mixed form in comparison to the control values. During remission, the levels of NO metabolites in the urine remain increased in both groups. In relapse of SSNS arginase levels in the urine increased by 4,5 times in comparation to SRNS, thus the concentration of arginase was reduced. During remission period arginase levels in the urine were practically reduced to the levels of the control group. In the mixed form of CGN, relapse period arginase levels in the urine were increased by 2,9 times and during remission were decreased by almost 1,9 times in comparision to the control group. CONCLUSIONS Assesment of NO metabolites and arginase in urine can be used as a diagnostic method in order to monitor renal disease process, evolution and effectiveness of the applied treatment.
Collapse
Affiliation(s)
- A Ciuntu
- Pediatric Department, State University of Medicine and Pharmacy "Nicolae Testemitanu", Chișinău Paediatrics National Institute of Health Care for Mother and Child, Nephrology Unit, R.Moldova
| |
Collapse
|
48
|
Dellera F, Ganzetti GS, Froio A, Manzini S, Busnelli M, Meinitzer A, Sirtori CR, Chiesa G, Parolini C. L-homoarginine administration reduces neointimal hyperplasia in balloon-injured rat carotids. Thromb Haemost 2016; 116:400-2. [PMID: 27279573 DOI: 10.1160/th15-10-0831] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 04/30/2016] [Indexed: 12/17/2022]
Abstract
Supplementary Material to this article is available at www.thrombosis-online.com.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Cinzia Parolini
- Cinzia Parolini, PhD, Department of Pharmacological and Biomolecular Sciences, Via Balzaretti 9, 20133 Milano, Italy, Tel.: +39 02 50318328, Fax: +39 02-50318284, E-mail:
| |
Collapse
|
49
|
Resveratrol improves vasoprotective effects of captopril on aortic remodeling and fibrosis triggered by renovascular hypertension. Cardiovasc Pathol 2016; 25:116-9. [DOI: 10.1016/j.carpath.2015.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 11/03/2015] [Accepted: 11/07/2015] [Indexed: 11/20/2022] Open
|
50
|
Nara A, Nagai H, Shintani-Ishida K, Ogura S, Shimosawa T, Kuwahira I, Shirai M, Yoshida KI. Pulmonary arterial hypertension in rats due to age-related arginase activation in intermittent hypoxia. Am J Respir Cell Mol Biol 2015; 53:184-92. [PMID: 25490411 DOI: 10.1165/rcmb.2014-0163oc] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is prevalent in patients with obstructive sleep apnea syndrome (OSAS). Aging induces arginase activation and reduces nitric oxide (NO) production in the arteries. Intermittent hypoxia (IH), conferred by cycles of brief hypoxia and normoxia, contributes to OSAS pathogenesis. Here, we studied the role of arginase and aging in the pathogenesis of PAH in adult (9-mo-old) and young (2-mo-old) male Sprague-Dawley rats subjected to IH or normoxia for 4 weeks and analyzed them with a pressure-volume catheter inserted into the right ventricle (RV) and by pulsed Doppler echocardiography. Western blot analysis was conducted on arginase, NO synthase isoforms, and nitrotyrosine. IH induced PAH, as shown by increased RV systolic pressure and RV hypertrophy, in adult rats but not in young rats. IH increased expression levels of arginase I and II proteins in the adult rats. IH also increased arginase I expression in the pulmonary artery endothelium and arginase II in the pulmonary artery adventitia. Furthermore, IH reduced pulmonary levels of nitrate and nitrite but increased nitrotyrosine levels in adult rats. An arginase inhibitor (N(ω)-hydroxy-nor-1-arginine) prevented IH-induced PAH and normalized nitrite and nitrate levels in adult rats. IH induced arginase up-regulation and PAH in adult rats, but not in young rats, through reduced NO production. Our findings suggest that arginase inhibition prevents or reverses PAH.
Collapse
Affiliation(s)
- Akina Nara
- 1 Department of Forensic Medicine, Graduate School of Medicine and
| | - Hisashi Nagai
- 1 Department of Forensic Medicine, Graduate School of Medicine and
| | | | - Sayoko Ogura
- 1 Department of Forensic Medicine, Graduate School of Medicine and.,2 Division of Laboratory Medicine, Department of Pathology and Microbiology, Faculty of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Tatsuo Shimosawa
- 3 Department of Clinical Laboratory, Faculty of Medicine, the University of Tokyo, Tokyo, Japan
| | - Ichiro Kuwahira
- 4 Department of Respiratory Medicine, Tokai University School of Medicine, Kanagawa, Japan; and
| | - Mikiyasu Shirai
- 5 Department of Cardiac Physiology, National Cerebral & Cardiovascular Center, Osaka, Japan
| | - Ken-ichi Yoshida
- 1 Department of Forensic Medicine, Graduate School of Medicine and
| |
Collapse
|