1
|
Dugbartey GJ. Nitric oxide in kidney transplantation. Biomed Pharmacother 2023; 167:115530. [PMID: 37722191 DOI: 10.1016/j.biopha.2023.115530] [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: 06/12/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 09/20/2023] Open
Abstract
Kidney transplantation is the treatment of choice for patients with kidney failure. Compared to dialysis therapy, it provides better quality of life and confers significant survival advantage at a relatively lower cost. However, the long-term success of this life-saving intervention is severely hampered by an inexorable clinical problem referred to as ischemia-reperfusion injury (IRI), and increases the incidence of post-transplant complications including loss of renal graft function and death of transplant recipients. Burgeoning evidence shows that nitric oxide (NO), a poisonous gas at high concentrations, and with a historic negative public image as an environmental pollutant, has emerged as a potential candidate that holds clinical promise in mitigating IRI and preventing acute and chronic graft rejection when it is added to kidney preservation solutions at low concentrations or when administered to the kidney donor prior to kidney procurement and to the recipient or to the reperfusion circuit at the start and during reperfusion after renal graft preservation. Interestingly, dysregulated or abnormal endogenous production and metabolism of NO is associated with IRI in kidney transplantation. From experimental and clinical perspectives, this review presents endogenous enzymatic production of NO as well as its exogenous sources, and then discusses protective effects of constitutive nitric oxide synthase (NOS)-derived NO against IRI in kidney transplantation via several signaling pathways. The review also highlights a few isolated studies of renal graft protection by NO produced by inducible NOS.
Collapse
Affiliation(s)
- George J Dugbartey
- Department of Pharmacology and Toxicology, School of Pharmacy, College of Health Sciences, University of Ghana, Legon, Accra, Ghana; Accra College of Medicine, Magnolia St, JVX5+FX9, East Legon, Accra, Ghana.
| |
Collapse
|
2
|
Sunga GM, Hartgerink J, Sikora AG, Young S. Enhancement of Immunotherapies in Head and Neck Cancers Using Biomaterial-Based Treatment Strategies. Tissue Eng Part C Methods 2023; 29:257-275. [PMID: 37183412 PMCID: PMC10282827 DOI: 10.1089/ten.tec.2023.0090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/12/2023] [Indexed: 05/16/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is a challenging disease to treat because of typically late-stage diagnoses and tumor formation in difficult-to-treat areas, sensitive to aggressive or invasive treatments. To date, HNSCC treatments have been limited to surgery, radiotherapy, and chemotherapy, which may have significant morbidity and often lead to long-lasting side effects. The development of immunotherapies has revolutionized cancer treatment by providing a promising alternative to standard-of-care therapies. However, single-agent immunotherapy has been only modestly effective in the treatment of various cancers, including HNSCC, with most patients receiving no overall benefit or increased survival. In addition, single-agent immunotherapy's limitations, namely immune-related side effects and the necessity of multidose treatments, must be addressed to further improve treatment efficacy. Biocompatible biomaterials, in combination with cancer immunotherapies, offer numerous advantages in the concentration, localization, and controlled release of drugs, cancer antigens, and immune cells. Biomaterial structures are diverse, and their design can generally be customized to enhance immunotherapy response. In preclinical settings, the use of biomaterials has shown great promise in improving the efficacy of single-agent immunotherapy. Herein, we provide an overview of current immunotherapy treatments for HNSCC and their limitations, as well as the potential applications of biomaterials in enhancing cancer immunotherapies. Impact Statement Advances in anticancer immunotherapies for the past 30 years have yielded exciting clinical results and provided alternatives to long-standing standard-of-care treatments, which are associated with significant toxicities and long-term morbidity. However, patients with head and neck squamous cell carcinoma (HNSCC) have not benefited from immunotherapies as much as patients with other cancers. Immunotherapy limitations include systemic side effects, therapeutic resistance, poor delivery kinetics, and limited patient responses. Biomaterial-enhanced immunotherapies, as explored in this review, are a potentially powerful means of achieving localized drug delivery, sustained and controlled drug release, and immunomodulation. They may overcome current treatment limitations and improve patient outcomes and care.
Collapse
Affiliation(s)
- Gemalene M. Sunga
- Katz Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Jeffrey Hartgerink
- Department of Chemistry, Rice University, Houston, Texas, USA
- Department of Bioengineering, Rice University, Houston, Texas, USA
| | - Andrew G. Sikora
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Simon Young
- Katz Department of Oral and Maxillofacial Surgery, The University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| |
Collapse
|
3
|
Ruan B, Chen Y, Trimidal S, Koo I, Qian F, Cai J, Mcguigan J, Hall MA, Patterson AD, Prabhu KS, Paulson RF. Nitric oxide regulates metabolism in murine stress erythroid progenitors to promote recovery during inflammatory anemia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.11.532207. [PMID: 36945370 PMCID: PMC10028999 DOI: 10.1101/2023.03.11.532207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
Inflammation skews bone marrow hematopoiesis increasing the production of myeloid effector cells at the expense of steady-state erythropoiesis. A compensatory stress erythropoiesis response is induced to maintain homeostasis until inflammation is resolved. In contrast to steady-state erythroid progenitors, stress erythroid progenitors (SEPs) utilize signals induced by inflammatory stimuli. However, the mechanistic basis for this is not clear. Here we reveal a nitric oxide (NO)-dependent regulatory network underlying two stages of stress erythropoiesis, namely proliferation, and the transition to differentiation. In the proliferative stage, immature SEPs and cells in the niche increased expression of inducible nitric oxide synthase ( Nos2 or iNOS ) to generate NO. Increased NO rewires SEP metabolism to increase anabolic pathways, which drive the biosynthesis of nucleotides, amino acids and other intermediates needed for cell division. This NO-dependent metabolism promotes cell proliferation while also inhibiting erythroid differentiation leading to the amplification of a large population of non-committed progenitors. The transition of these progenitors to differentiation is mediated by the activation of nuclear factor erythroid 2-related factor 2 (Nfe2l2 or Nrf2). Nrf2 acts as an anti-inflammatory regulator that decreases NO production, which removes the NO-dependent erythroid inhibition and allows for differentiation. These data provide a paradigm for how alterations in metabolism allow inflammatory signals to amplify immature progenitors prior to differentiation. Key points Nitric-oxide (NO) dependent signaling favors an anabolic metabolism that promotes proliferation and inhibits differentiation.Activation of Nfe2l2 (Nrf2) decreases NO production allowing erythroid differentiation.
Collapse
|
4
|
Ghonimy A, Chen Z, Li J. The effect of C/N ratio and its frequent addition on commensal and pathogenic bacterial abundances in shrimp Litopeaneus vanname gut in a biofloc system: Ratio and frequent addition interaction matters. PLoS One 2023; 18:e0283841. [PMID: 37011061 PMCID: PMC10069773 DOI: 10.1371/journal.pone.0283841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 03/19/2023] [Indexed: 04/05/2023] Open
Abstract
The environmental biotic and abiotic factors form a complicated relationship with the host intestinal microbiota. In our study, we applied different levels of C/N ratio (10, 15, 20) and frequent addition times (once, twice, triple a day) in a factorial experimental design. GC/LC analysis of filtrated biofloc (BF) samples revealed the highest relative fold change for the untargeted bioactive molecules among different treatments, whereas the 16s rRNA analysis revealed the change in the shrimp gut microbiota composition. Based on the available literature on the relationship between the bioactive molecules and the available bacteria in this study, the next bioactive molecules were discussed. Proline was associated with Bacteroidota, Flavobacteriaceae, Gammaproteobacteria, and Flavobacteriales. Plumbagine was associated with Norcardiaceae. Phytosphingosin was associated with Bacteroidota. Phosphocholine compound was associated with Bacteroidota. The monobutyl ether, benzofuran, and piperidone were associated with Micobacteriaceae and Mycobacterium. Generally, C/N 15 and 20 once a day, and C/N 20 triple a day have showed a merit over other treatments in term of low pathogenic and unfavorable bacteria, and high commensal bacterial abundances. The revealed bioactive molecule composition showed the complicity of BF as a source for novel compounds as biosecurity agents in BF system. These molecules could be developed to feed additives upgrading the biosecurity level in aquaculture systems. Other bioactive molecules require future studies to reveal novel molecules in term of aquaculture biosecurity control.
Collapse
Affiliation(s)
- Abdallah Ghonimy
- Key Laboratory of Sustainable Development of Marine Fisheries, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Zhao Chen
- Key Laboratory of Sustainable Development of Marine Fisheries, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| | - Jian Li
- Key Laboratory of Sustainable Development of Marine Fisheries, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
| |
Collapse
|
5
|
Yin YL, Chen Y, Ren F, Wang L, Zhu ML, Lu JX, Wang QQ, Lu CB, Liu C, Bai YY, Wang SX, Wang JZ, Li P. Nitrosative stress induced by homocysteine thiolactone drives vascular cognitive impairments via GTP cyclohydrolase 1 S-nitrosylation in vivo. Redox Biol 2022; 58:102540. [DOI: 10.1016/j.redox.2022.102540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/02/2022] [Accepted: 11/11/2022] [Indexed: 11/15/2022] Open
|
6
|
Rotoli BM, Visigalli R, Ferrari F, Ranieri M, Tamma G, Dall’Asta V, Barilli A. Desmopressin Stimulates Nitric Oxide Production in Human Lung Microvascular Endothelial Cells. Biomolecules 2022; 12:biom12030389. [PMID: 35327581 PMCID: PMC8945551 DOI: 10.3390/biom12030389] [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: 01/12/2022] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 01/10/2023] Open
Abstract
Desmopressin (dDAVP) is the best characterized analogue of vasopressin, the endocrine regulator of water balance endowed with potent vasoconstrictive effects. Despite the use of dDAVP in clinical practice, ranging from the treatment of nephrogenic diabetes insipidus to bleeding disorders, much remains to be understood about the impact of the drug on endothelial phenotype. The aim of this study was, thus, to evaluate the effects of desmopressin on the viability and function of human pulmonary microvascular endothelial cells (HLMVECs). The results obtained demonstrate that the vasopressor had no cytotoxic effect on the endothelium; similarly, no sign of endothelial activation was induced by dDAVP, indicated by the lack of effect on the expression of inflammatory cytokines and adhesion molecules. Conversely, the drug significantly stimulated the production of nitric oxide (NO) and the expression of the inducible isoform of nitric oxide synthase, NOS2/iNOS. Since the intracellular level of cAMP also increased, we can hypothesize that NO release is consequent to the activation of the vasopressin receptor 2 (V2R)/guanylate cyclase (Gs)/cAMP axis. Given the multifaceted role of NOS2-deriving NO for many physio-pathological conditions, the meanings of these findings in HLMVECs appears intriguing and deserves to be further addressed.
Collapse
Affiliation(s)
- Bianca Maria Rotoli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (B.M.R.); (R.V.); (F.F.); (V.D.)
| | - Rossana Visigalli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (B.M.R.); (R.V.); (F.F.); (V.D.)
| | - Francesca Ferrari
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (B.M.R.); (R.V.); (F.F.); (V.D.)
| | - Marianna Ranieri
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (M.R.); (G.T.)
| | - Grazia Tamma
- Department of Bioscience, Biotechnology and Biopharmaceutics, University of Bari, 70125 Bari, Italy; (M.R.); (G.T.)
| | - Valeria Dall’Asta
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (B.M.R.); (R.V.); (F.F.); (V.D.)
| | - Amelia Barilli
- Laboratory of General Pathology, Department of Medicine and Surgery, University of Parma, 43125 Parma, Italy; (B.M.R.); (R.V.); (F.F.); (V.D.)
- Correspondence:
| |
Collapse
|
7
|
Suga Y, Takahashi Y, Shimada T, Yamada S, Morishita E, Asakura H. Effect of NOS Inhibitors and Anticoagulants on Nitric Oxide Production in a Tissue-factor Induced Rat DIC Model. In Vivo 2021; 35:1999-2004. [PMID: 34182474 DOI: 10.21873/invivo.12468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM We examined the mechanism of nitric oxide (NO) production in a tissue-factor (TF)-induced disseminated intravascular coagulation (DIC) model in rats, using inducible nitric oxide synthase (iNOS) inhibitor (L-NIL), endothelial nitric oxide synthase (eNOS) inhibitor (L-NAME), Factor Xa inhibitor (DX-9065a), and thrombin inhibitor argatroban. MATERIALS AND METHODS Experimental DIC was induced by sustained infusion of 3.75 U/kg TF for 4 h via the tail vein. We then investigated the effect of these four agents on TF-induced DIC. RESULTS Administration of L-NIL or L-NAME during induction of TF-induced DIC did not affect hemostatic markers, whereas elevated plasma levels of NO metabolites (NOX) were significantly suppressed by co-administration of L-NAME. A significant increase in eNOS-mRNA expression was observed in the TF-induced DIC model. Argatroban almost completely suppressed eNOS-mRNA expression. CONCLUSION eNOS plays an important role in the NO production in the TF-induced DIC, and thrombin is a key stimulant of eNOS-mRNA expression in this model.
Collapse
Affiliation(s)
- Yukio Suga
- Department of Clinical Pharmacy and Healthcare Science, Faculty of Pharmacy, Institute of Medical, Pharmaceutical & Health Science, Kanazawa University, Kanazawa, Japan;
| | - Yoko Takahashi
- Department of Clinical Pharmacy and Healthcare Science, Faculty of Pharmacy, Institute of Medical, Pharmaceutical & Health Science, Kanazawa University, Kanazawa, Japan
| | - Tsutomu Shimada
- Department of Hospital Pharmacy, University Hospital, Kanazawa University, Kanazawa, Japan
| | - Shinya Yamada
- Department of Hematology, Kanazawa University Hospital, Kanazawa, Japan
| | - Eriko Morishita
- Department of Hematology, Kanazawa University Hospital, Kanazawa, Japan
| | - Hidesaku Asakura
- Department of Hematology, Kanazawa University Hospital, Kanazawa, Japan
| |
Collapse
|
8
|
Chen S, Guo C, Wang R, Feng Z, Liu Z, Wu L, Zhao D, Zheng S, Chen F, Zhang D, Xu J, Zhu J, Chen X, Li Z, Wise CM, Li J, Wang XY. Monocytic MDSCs skew Th17 cells toward a pro-osteoclastogenic phenotype and potentiate bone erosion in rheumatoid arthritis. Rheumatology (Oxford) 2021; 60:2409-2420. [PMID: 33246326 DOI: 10.1093/rheumatology/keaa625] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 08/24/2020] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVES While myeloid-derived suppressor cells (MDSCs) were previously shown to promote a proinflammatory T helper (Th) 17 response in autoimmune conditions, a potential impact of the MDSC-Th17 immune axis on abnormal bone destruction in RA remains largely unknown. METHODS We investigated the correlation between the frequency of MDSCs or its subsets and joint destruction in RA patients. The reciprocal actions of patient-derived MDSCs and Th17 cells were studied using osteoclast (OC) differentiation and bone resorption assays in vitro, which were further validated using mouse models of RA. Contribution of MDSCs to osteoclastogenesis and bone erosion in vivo was determined by depletion or transfer of MDSCs. RESULTS Human MDSCs, particularly monocytic MDSCs (M-MDSCs), exhibit inherent OC-differentiating capacity and positively correlate with clinical bone erosion in RA patients. Strikingly, patient-derived M-MDSCs can program Th17 cells towards a pro-osteoclastogenic phenotype, which in return potentiates OC differentiation via the receptor activator of nuclear factor κΒ ligand (RANK-L)-RANK signalling. This enhanced osteolysis driven by the reciprocal actions of M-MDSCs and Th17 cells is further confirmed using mouse models of RA. Selective depletion of M-MDSCs significantly ameliorates osteoclastogenesis and disease severity in arthritic mice, whereas transfer of M-MDSCs aggravates bone erosion associated with increased OCs in recipient mice. CONCLUSION Our findings highlight the functional plasticity of MDSCs and identify a novel pro-osteoclastogenic pathway governed by interplay between myeloid cells and T lymphocytes in autoimmune RA.
Collapse
Affiliation(s)
- Shixian Chen
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine.,Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunqing Guo
- Department of Human & Molecular Genetics.,Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Ran Wang
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine.,Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhitao Feng
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine
| | - Zheng Liu
- Department of Human & Molecular Genetics.,Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Lisheng Wu
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine
| | - Di Zhao
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine
| | - Songyuan Zheng
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine
| | - Feilong Chen
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine
| | - Dingding Zhang
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine
| | - Juan Xu
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine
| | - Junqing Zhu
- Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoguang Chen
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Pathogen Biology, Southern Medical University School of Public Health, Guangzhou
| | - Zhanguo Li
- Department of Rheumatology and Immunology, Peking University People's Hospital & Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis, Beijing, China
| | - Christopher M Wise
- Department of Internal Medicine, Virginia Commonwealth University Medical Center
| | - Juan Li
- Department of Traditional Chinese Internal Medicine, School of Traditional Chinese Medicine.,Department of Rheumatic & TCM Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiang-Yang Wang
- Department of Human & Molecular Genetics.,Institute of Molecular Medicine, Virginia Commonwealth University School of Medicine, Richmond, VA, USA.,McGuire VA Medical Center, Richmond, VA, USA
| |
Collapse
|
9
|
Sassetti E, Clausen MH, Laraia L. Small-Molecule Inhibitors of Reactive Oxygen Species Production. J Med Chem 2021; 64:5252-5275. [PMID: 33856791 DOI: 10.1021/acs.jmedchem.0c01914] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are involved in physiological cellular processes including differentiation, proliferation, and apoptosis by acting as signaling molecules or regulators of transcription factors. The maintenance of appropriate cellular ROS levels is termed redox homeostasis, a balance between their production and neutralization. High concentrations of ROS may contribute to severe pathological events including cancer, neurodegenerative, and cardiovascular diseases. In recent years, approaches to target the sources of ROS production directly in order to develop tool compounds or potential therapeutics have been explored. Herein, we briefly outline the major sources of cellular ROS production and comprehensively review the targeting of these by small-molecule inhibitors. We critically assess the value of ROS inhibitors with different mechanisms-of-action, including their potency, mode-of-action, known off-target effects, and clinical or preclinical status, while suggesting future avenues of research in the field.
Collapse
Affiliation(s)
- Elisa Sassetti
- Center for Nanomedicine and Theranostics, Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark
| | - Mads H Clausen
- Center for Nanomedicine and Theranostics, Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark
| | - Luca Laraia
- Center for Nanomedicine and Theranostics, Department of Chemistry, Technical University of Denmark, Kemitorvet 207, 2800 Kgs. Lyngby, Denmark
| |
Collapse
|
10
|
Murakawa-Hirachi T, Mizoguchi Y, Ohgidani M, Haraguchi Y, Monji A. Effect of memantine, an anti-Alzheimer's drug, on rodent microglial cells in vitro. Sci Rep 2021; 11:6151. [PMID: 33731780 PMCID: PMC7969939 DOI: 10.1038/s41598-021-85625-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 02/24/2021] [Indexed: 01/07/2023] Open
Abstract
The pathophysiology of Alzheimer's disease (AD) is related to neuroinflammatory responses mediated by microglia. Memantine, an antagonist of N-methyl-D-aspartate (NMDA) receptors used as an anti-Alzheimer's drug, protects from neuronal death accompanied by suppression of proliferation and activation of microglial cells in animal models of AD. However, it remains to be tested whether memantine can directly affect microglial cell function. In this study, we examined whether pretreatment with memantine affects intracellular NO and Ca2+ mobilization using DAF-2 and Fura-2 imaging, respectively, and tested the effects of memantine on phagocytic activity by human β-Amyloid (1-42) phagocytosis assay in rodent microglial cells. Pretreatment with memantine did not affect production of NO or intracellular Ca2+ elevation induced by TNF in rodent microglial cells. Pretreatment with memantine also did not affect the mRNA expression of pro-inflammatory (TNF, IL-1β, IL-6 and CD45) or anti-inflammatory (IL-10, TGF-β and arginase) phenotypes in rodent microglial cells. In addition, pretreatment with memantine did not affect the amount of human β-Amyloid (1-42) phagocytosed by rodent microglial cells. Moreover, we observed that pretreatment with memantine did not affect 11 major proteins, which mainly function in the phagocytosis and degradation of β-Amyloid (1-42), including TREM2, DAP12 and neprilysin in rodent microglial cells. To the best of our knowledge, this is the first report to suggest that memantine does not directly modulate intracellular NO and Ca2+ mobilization or phagocytic activity in rodent microglial cells. Considering the neuroinflammation hypothesis of AD, the results might be important to understand the effect of memantine in the brain.
Collapse
Affiliation(s)
- Toru Murakawa-Hirachi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Yoshito Mizoguchi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan.
| | - Masahiro Ohgidani
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
- Department of Integrative Anatomy, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshinori Haraguchi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Akira Monji
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| |
Collapse
|
11
|
Leach DG, Dharmaraj N, Lopez-Silva TL, Venzor JR, Pogostin BH, Sikora AG, Hartgerink JD, Young S. Biomaterial-Facilitated Immunotherapy for Established Oral Cancers. ACS Biomater Sci Eng 2021; 7:415-421. [PMID: 33470801 PMCID: PMC8325389 DOI: 10.1021/acsbiomaterials.0c01575] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We evaluated a peptide-based immunotherapy termed SynerGel: an injectable, biomaterial-based platform for intratumoral drug delivery. A drug-mimicking peptide hydrogel named L-NIL-MDP was loaded with an antitumor cyclic dinucleotide (CDN) immunotherapy agonist. The biomaterial combines inducible nitric oxide synthase (iNOS) inhibition with controlled delivery of CDNs, demonstrating between 4- and 20-fold slower drug release than commercially available hydrogels. SynerGel allowed for immune-mediated elimination of established treatment-resistant oral tumors in a murine model, with a median survival of 67.5 days compared with 44 days in no-treatment control. This report details findings for a promising therapy showing improved efficacy over previous hydrogel systems.
Collapse
Affiliation(s)
- David G. Leach
- Department of Chemistry, Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Neeraja Dharmaraj
- Department of Oral & Maxillofacial Surgery, The University of Texas Health Science Center at Houston School of Dentistry, TX 77054, USA
| | - Tania L. Lopez-Silva
- Department of Chemistry, Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Jose Rodriguez Venzor
- Department of Oral & Maxillofacial Surgery, The University of Texas Health Science Center at Houston School of Dentistry, TX 77054, USA
| | - Brett H. Pogostin
- Department of Chemistry, Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Andrew G. Sikora
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeffrey D. Hartgerink
- Department of Chemistry, Department of Bioengineering, Rice University, Houston, TX 77005, USA
| | - Simon Young
- Department of Oral & Maxillofacial Surgery, The University of Texas Health Science Center at Houston School of Dentistry, TX 77054, USA
| |
Collapse
|
12
|
Ali EMH, Abdel-Maksoud MS, Hassan RM, Mersal KI, Ammar UM, Se-In C, He-Soo H, Kim HK, Lee A, Lee KT, Oh CH. Design, synthesis and anti-inflammatory activity of imidazol-5-yl pyridine derivatives as p38α/MAPK14 inhibitor. Bioorg Med Chem 2021; 31:115969. [PMID: 33422910 DOI: 10.1016/j.bmc.2020.115969] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 01/12/2023]
Abstract
P38α/MAPK14 is intracellular signalling regulator involved in biosynthesis of inflammatory mediator cytokines (TNF-α, IL-1, IL-6, and IL-1b), which induce the production of inflammatory proteins (iNOS, NF-kB, and COX-2). In this study, drug repurposing strategies were followed to repositioning of a series of B-RAF V600E imidazol-5-yl pyridine inhibitors to inhibit P38α kinase. A group 25 reported P38α kinase inhibitors were used to build a pharmacophore model for mapping the target compounds and proving their affinity for binding in P38α active site. Target compounds were evaluated for their potency against P38α kinase, compounds 11a and 11d were the most potent inhibitors (IC50 = 47 nM and 45 nM, respectively). In addition, compound 11d effectively inhibited the production of proinflammatory cytokinesTNF-α, 1L-6, and 1L-1β in LPS-induced RAW 264.7 macrophages with IC50 values of 78.03 nM, 17.6 µM and 82.15 nM, respectively. The target compounds were tested for their anti-inflammatory activity by detecting the reduction of Nitric oxide (NO) and prostaglandin (PGE2) production in LPS-stimulated RAW 264.7 macrophages. Compound 11d exhibited satisfied inhibitory activity of the production of PGE2 and NO with IC50 values of 0.29 µM and 0.61 µM, respectively. Molecular dynamics simulations of the most potent inhibitor 11d were carried out to illustrate its conformational stability in the binding site of P38α kinase.
Collapse
Affiliation(s)
- Eslam M H Ali
- Center for Biomaterials, Korea Institute of Science & Technology (KIST School), Seoul, Seongbuk-gu, 02792, Republic of Korea; University of Science & Technology (UST), Daejeon, Yuseong-gu, 34113, Republic of Korea; Pharmaceutical Chemistry Department, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo 12055, Egypt
| | - Mohammed S Abdel-Maksoud
- Medicinal & Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre (NRC), (ID: 60014618), P.O. 12622, Dokki, Giza, Egypt
| | - Rasha Mohamed Hassan
- Medicinal & Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre (NRC), (ID: 60014618), P.O. 12622, Dokki, Giza, Egypt
| | - Karim I Mersal
- Center for Biomaterials, Korea Institute of Science & Technology (KIST School), Seoul, Seongbuk-gu, 02792, Republic of Korea; University of Science & Technology (UST), Daejeon, Yuseong-gu, 34113, Republic of Korea
| | - Usama M Ammar
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0NR, Scotland, United Kingdom
| | - Choi Se-In
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Han He-Soo
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | - Hee-Kwon Kim
- Department of Nuclear Medicine, Molecular Imaging & Therapeutic Medicine Research Center, Jeonbuk National University Medical School and Hospital, 20 Geonji-ro, Deokjin-gu, Jeonju 54907, Republic of Korea; Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, 20 Geonji-ro, Deokjin-gu, Jeonju 54907, Republic of Korea
| | - Anna Lee
- Department of Chemistry, Hanseo University, Seosan 31962, Republic of Korea
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, 1 Hoegi-dong, Dongdaemun-gu, Seoul 130-701, Republic of Korea.
| | - Chang-Hyun Oh
- Center for Biomaterials, Korea Institute of Science & Technology (KIST School), Seoul, Seongbuk-gu, 02792, Republic of Korea; University of Science & Technology (UST), Daejeon, Yuseong-gu, 34113, Republic of Korea.
| |
Collapse
|
13
|
DeAdder NP, Gillam HJ, Wilson BC. Relaxin peptides reduce cellular damage in cultured brain slices exposed to transient oxygen–glucose deprivation: an effect mediated by nitric oxide. Facets (Ott) 2021. [DOI: 10.1139/facets-2020-0029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The effect of treatment with human relaxins on cell death was studied in oxygen- and glucose-deprived brain slices. In addition, involvement of nitric oxide and the relaxin receptor, RXFP3, was studied. Brain slices ( n = 12–18/group) were cultured under standard conditions for two weeks and then exposed to: ( i) an oxygenated balanced salt solution, ( ii) a deoxygenated, glucose-free balanced salt solution (OGD media), or ( iii) OGD media containing 10−7 mol/L H2 relaxin, 10−7 mol/L H2 relaxin with 50 μmol/L L-NIL, 10−7 mol/L H3 relaxin, or 10−7 mol/L H3 relaxin with 50 μmol/L L-NIL. Cell death was assessed using propidium iodide fluorescence. In a separate experiment, 10−5 mol/L R3 B1-22R (an antagonist of RXFP3) was added to both H2 and H3 relaxin treatments. H2 and H3 relaxin treatment reduced cell damage or death in OGD slices and L-NIL partially attenuated the effect of H3 relaxin. Antagonism of RXFP3 blocked the effect of H3 but not H2 relaxin. These data increase our understanding of the role of relaxin ligands and their receptors in protecting tissues throughout the body from ischemia and reperfusion injury.
Collapse
Affiliation(s)
| | - Hannah J. Gillam
- Department of Biology, Acadia University, Wolfville, NS B4P 2R6, Canada
| | - Brian C. Wilson
- Department of Biology, Acadia University, Wolfville, NS B4P 2R6, Canada
| |
Collapse
|
14
|
Limiting oxidative DNA damage reduces microbe-induced colitis-associated colorectal cancer. Nat Commun 2020; 11:1802. [PMID: 32286276 PMCID: PMC7156452 DOI: 10.1038/s41467-020-15549-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 03/17/2020] [Indexed: 12/31/2022] Open
Abstract
Inflammatory bowel disease patients have a greatly increased risk of developing colitis-associated colon cancer (CAC); however, the basis for inflammation-induced genetic damage requisite for neoplasia is unclear. Using three models of CAC, we find that sustained inflammation triggers 8-oxoguanine DNA lesions. Strikingly, antioxidants or iNOS inhibitors reduce 8-oxoguanine and polyps in CAC models. Because the mismatch repair (MMR) system repairs 8-oxoguanine and is frequently defective in colorectal cancer (CRC), we test whether 8-oxoguanine mediates oncogenesis in a Lynch syndrome (MMR-deficient) model. We show that microbiota generates an accumulation of 8-oxoguanine lesions in MMR-deficient colons. Accordingly, we find that 8-oxoguanine is elevated in neoplastic tissue of Lynch syndrome patients compared to matched untransformed tissue or non-Lynch syndrome neoplastic tissue. While antioxidants reduce 8-oxoguanine, they do not reduce CRC in Lynch syndrome models. Hence, microbe-induced oxidative/nitrosative DNA damage play causative roles in inflammatory CRC models, but not in Lynch syndrome models. It is unclear how microbial-induced inflammation promotes neoplastic transformation in colitis-associated cancer (CAC). Here, the authors use models of CAC to show that inflammation induces 8-oxoguanine lesions in DNA, and that antioxidants can reduce these DNA lesions as well as CAC.
Collapse
|
15
|
Mittal A, Kakkar R. Nitric Oxide Synthases and Their Inhibitors: A Review. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180816666190222154457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Nitric Oxide (NO), an important biological mediator, is involved in the regulation of the cardiovascular, nervous and immune systems in mammals. Synthesis of NO is catalyzed by its biosynthetic enzyme, Nitric Oxide Synthase (NOS). There are three main isoforms of the enzyme, neuronal NOS, endothelial NOS and inducible NOS, which have very similar structures but differ in their expression and activities. NO is produced in the active site of the enzyme in two distinct cycles from oxidation of the substrate L-arg (L-arginine) in nicotinamide adenine dinucleotide phosphate (NADPH)-dependent reaction. NOS has gained considerable attention of biochemists due to its complexity and unique catalytic mechanism. The review focuses on NOS structure, its function and catalytic reaction mechanism. In particular, the review is concluded with a discussion on the role of all three isoforms of NOS in physiological and pathological conditions and their inhibitors with a focus on the role of computational techniques in their development.
Collapse
Affiliation(s)
- Anshika Mittal
- Computational Chemistry Laboratory, Department of Chemistry, University of Delhi, Delhi-110007, India
| | - Rita Kakkar
- Computational Chemistry Laboratory, Department of Chemistry, University of Delhi, Delhi-110007, India
| |
Collapse
|
16
|
Touati-Jallabe Y, Tintillier T, Mauchauffée E, Boucher JL, Leroy J, Ramassamy B, Hamzé A, Mezghenna K, Bouzekrini A, Verna C, Martinez J, Lajoix AD, Hernandez JF. Solid-Phase Synthesis of Substrate-Based Dipeptides and Heterocyclic Pseudo-dipeptides as Potential NO Synthase Inhibitors. ChemMedChem 2020; 15:517-531. [PMID: 32027778 DOI: 10.1002/cmdc.201900659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/03/2020] [Indexed: 11/06/2022]
Abstract
More than 160 arginine analogues modified on the C-terminus via either an amide bond or a heterocyclic moiety (1,2,4-oxadiazole, 1,3,4-oxadiazole and 1,2,4-triazole) were prepared as potential inhibitors of NO synthases (NOS). A methodology involving formation of a thiocitrulline intermediate linked through its side-chain on a solid support followed by modification of its carboxylate group was developed. Finally, the side-chain thiourea group was either let unchanged, S-alkylated (Me, Et) or guanidinylated (Me, Et) to yield respectively after TFA treatment the corresponding thiocitrulline, S-Me/Et-isothiocitrulline and N-Me/Et-arginine substrate analogues. They all were tested against three recombinant NOS isoforms. Several compounds containing a S-Et- or a S-Me-Itc moiety and mainly belonging to both the dipeptide-like and 1,2,4-oxadiazole series were shown to inhibit nNOS and iNOS with IC50 in the 1-50 μM range. Spectral studies confirmed that these new compounds interacted at the heme active site. The more active compounds were found to inhibit intra-cellular iNOS expressed in RAW264.7 and INS-1 cells with similar efficiency than the reference compounds L-NIL and SEIT.
Collapse
Affiliation(s)
- Youness Touati-Jallabe
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France.,Avara Pharmaceutical Services, Boucherville, QC, J4B 7 K8, Canada
| | - Thibault Tintillier
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France.,Asymptote Project Management, 1 rue Edisson, 69500, Bron, France
| | - Elodie Mauchauffée
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France
| | - Jean-Luc Boucher
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques UMR8601, CNRS, Université Paris-Descartes, 45 rue des Saints Pères, 75270, Paris Cedex 06, France
| | - Jérémy Leroy
- Centre Biocommunication en Cardio-métabolique, Université Montpellier, Faculté de Pharmacie, 34000, Montpellier, France
| | - Booma Ramassamy
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques UMR8601, CNRS, Université Paris-Descartes, 45 rue des Saints Pères, 75270, Paris Cedex 06, France
| | - Abdallah Hamzé
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France.,Current address: BioCIS, UMR 8076, CNRS, Université Paris Sud, Université Paris-Saclay, 92296, Châtenay-Malabry, France
| | - Karima Mezghenna
- Centre Biocommunication en Cardio-métabolique, Université Montpellier, Faculté de Pharmacie, 34000, Montpellier, France
| | - Amine Bouzekrini
- Centre Biocommunication en Cardio-métabolique, Université Montpellier, Faculté de Pharmacie, 34000, Montpellier, France
| | - Claudia Verna
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France
| | - Jean Martinez
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France
| | - Anne-Dominique Lajoix
- Centre Biocommunication en Cardio-métabolique, Université Montpellier, Faculté de Pharmacie, 34000, Montpellier, France
| | - Jean-François Hernandez
- Institut des Biomolécules Max Mousseron, Université Montpellier, CNRS, ENSCM, Faculté de Pharmacie, 34000, Montpellier, France
| |
Collapse
|
17
|
Cinelli MA, Do HT, Miley GP, Silverman RB. Inducible nitric oxide synthase: Regulation, structure, and inhibition. Med Res Rev 2020; 40:158-189. [PMID: 31192483 PMCID: PMC6908786 DOI: 10.1002/med.21599] [Citation(s) in RCA: 485] [Impact Index Per Article: 97.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/14/2019] [Accepted: 05/13/2019] [Indexed: 12/11/2022]
Abstract
A considerable number of human diseases have an inflammatory component, and a key mediator of immune activation and inflammation is inducible nitric oxide synthase (iNOS), which produces nitric oxide (NO) from l-arginine. Overexpressed or dysregulated iNOS has been implicated in numerous pathologies including sepsis, cancer, neurodegeneration, and various types of pain. Extensive knowledge has been accumulated about the roles iNOS plays in different tissues and organs. Additionally, X-ray crystal and cryogenic electron microscopy structures have shed new insights on the structure and regulation of this enzyme. Many potent iNOS inhibitors with high selectivity over related NOS isoforms, neuronal NOS, and endothelial NOS, have been discovered, and these drugs have shown promise in animal models of endotoxemia, inflammatory and neuropathic pain, arthritis, and other disorders. A major issue in iNOS inhibitor development is that promising results in animal studies have not translated to humans; there are no iNOS inhibitors approved for human use. In addition to assay limitations, both the dual modalities of iNOS and NO in disease states (ie, protective vs harmful effects) and the different roles and localizations of NOS isoforms create challenges for therapeutic intervention. This review summarizes the structure, function, and regulation of iNOS, with focus on the development of iNOS inhibitors (historical and recent). A better understanding of iNOS' complex functions is necessary before specific drug candidates can be identified for classical indications such as sepsis, heart failure, and pain; however, newer promising indications for iNOS inhibition, such as depression, neurodegenerative disorders, and epilepsy, have been discovered.
Collapse
Affiliation(s)
- Maris A. Cinelli
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
- Current address: Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824
| | - Ha T. Do
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
- Current address: Mersana Therapeutics, Inc., Cambridge, MA 02139
| | - Galen P. Miley
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
| | - Richard B. Silverman
- Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Center for Developmental Therapeutics, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
| |
Collapse
|
18
|
Leach DG, Newton JM, Florez MA, Lopez-Silva TL, Jones AA, Young S, Sikora AG, Hartgerink JD. Drug-Mimicking Nanofibrous Peptide Hydrogel for Inhibition of Inducible Nitric Oxide Synthase. ACS Biomater Sci Eng 2019; 5:6755-6765. [PMID: 33304997 DOI: 10.1021/acsbiomaterials.9b01447] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this work, we develop a drug-mimicking nanofibrous peptide hydrogel that shows long-term bioactivity comparable to a small-molecule inhibitor of inducible nitric oxide synthase (iNOS). The iNOS inhibitor, N 6-(1-iminoethyl)-l-lysine (l-NIL), is a positively charged amino acid whose structure could be readily integrated into the framework of a positively charged multidomain peptide (MDP) through the modification of lysine side chains. This new l-NIL-MDP maintains the self-assembling properties of the base peptide, forming β-sheet nanofibers, which entangle into a thixotropic hydrogel. The l-NIL-MDP hydrogel supports cell growth in vitro and allows syringe-directed delivery that persists in a targeted location in vivo for several weeks. Multiple characterization assays demonstrate the bioactivity of the l-NIL-MDP hydrogel to be comparable to the l-NIL small molecule. This includes iNOS inhibition of macrophages in vitro, reduced nitrotyrosine immunostaining in murine subcutaneous histology, and reduced serum levels of vascular endothelial growth factor in vivo. This study expands the toolbox of available peptide hydrogel scaffold designs that can modify biological activity without the need for any additional small-molecule drugs, proteins, or cells.
Collapse
Affiliation(s)
- David G Leach
- Department of Chemistry and Department of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Jared M Newton
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, Texas 77030, United States.,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Marcus A Florez
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, Texas 77030, United States.,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Tania L Lopez-Silva
- Department of Chemistry and Department of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Adrianna A Jones
- Department of Chemistry and Department of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Simon Young
- Department of Oral & Maxillofacial Surgery, University of Texas Health Science Center, Houston, Texas 77054, United States
| | - Andrew G Sikora
- Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Jeffrey D Hartgerink
- Department of Chemistry and Department of Bioengineering, Rice University, Houston, Texas 77005, United States
| |
Collapse
|
19
|
Hassan AH, Yoo SY, Lee KW, Yoon YM, Ryu HW, Jeong Y, Shin JS, Kang SY, Kim SY, Lee HH, Park BY, Lee KT, Lee YS. Repurposing mosloflavone/5,6,7-trimethoxyflavone-resveratrol hybrids: Discovery of novel p38-α MAPK inhibitors as potent interceptors of macrophage-dependent production of proinflammatory mediators. Eur J Med Chem 2019; 180:253-267. [DOI: 10.1016/j.ejmech.2019.07.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/21/2019] [Accepted: 07/08/2019] [Indexed: 12/17/2022]
|
20
|
Nitric oxide regulates the expression of heme carrier protein-1 via hypoxia inducible factor-1α stabilization. PLoS One 2019; 14:e0222074. [PMID: 31513628 PMCID: PMC6742216 DOI: 10.1371/journal.pone.0222074] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Accepted: 08/21/2019] [Indexed: 01/08/2023] Open
Abstract
Photodynamic therapy (PDT) is a cancer therapy that capitalizes on cancer-specific porphyrin accumulation. We have investigated this phenomenon to propose the following three conclusions: 1) the mechanism underlying this phenomenon is closely related to both nitric oxide (NO) and heme carrier protein-1 (HCP-1), 2) NO inactivates ferrochelatase, and thus, the intracellular porphyrin levels in the cells are increased by the administration of an NO donor after 5-aminolevulinic acid treatment, 3) HCP-1 transports not only heme but also other porphyrins. Since NO stabilizes hypoxia-inducible factor (HIF)-1α, resulting in the upregulation of heme biosynthesis, HCP-1 expression can be increased by HIF-1α stabilization. In this study, we determined whether NO regulates HCP-1 expression by stabilizing HIF-1α expression. For this purpose, rat gastric cancer cell line RGK36 was treated with L-arginine or N6-(1-iminoethyl)-L-lysine (L-NIL). L-arginine treatment increased the intracellular NO concentration, and both HCP-1 and HIF-1α expression, while L-NIL treatment decreased them. Cytotoxicity of PDT was enhanced by L-arginine, following intracellular hemato-porphyrin dihydrochloride (HpD) accumulation. Both Cytotoxicity of PDT and HpD accumulation were decreased by L-NIL. The HCP-1 and HIF-1α expression, intracellular HpD accumulation and PDT cytotoxicity were decreased by 2-methoxyestradiol, which is a HIF-1α inhibitor. Moreover, these phenomena were not increased by a combination of both L-arginine and 2-Me. Thus, HCP-1 can be a downstream target of HIF-1α. These effects were also induced in the human gastric cancer cell line MKN45. Taken together, we conclude that HCP-1 expression is regulated by NO via HIF-1α stabilization.
Collapse
|
21
|
Wanderley JLM, Deolindo P, Carlsen E, Portugal AB, DaMatta RA, Barcinski MA, Soong L. CD4 + T Cell-Dependent Macrophage Activation Modulates Sustained PS Exposure on Intracellular Amastigotes of Leishmania amazonensis. Front Cell Infect Microbiol 2019; 9:105. [PMID: 31032234 PMCID: PMC6473175 DOI: 10.3389/fcimb.2019.00105] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/26/2019] [Indexed: 01/16/2023] Open
Abstract
Leishmania amazonensis amastigotes can make use of surface-exposed phosphatidylserine (PS) molecules to promote infection and non-classical activation of macrophages (MΦ), leading to uncontrolled intracellular proliferation of the parasites. This mechanism was quoted as apoptotic mimicry. Moreover, the amount of PS molecules exposed on the surface of amastigotes correlates with the susceptibility of the host. In this study, we tested whether host cellular responses influence PS expression on intracellular amastigotes. We found that the level of PS exposure on intracellular amastigotes was modulated by CD4+ T cell and MΦ activation status in vitro and in vivo. L. amazonensis infection generated a Th1/Th2-mixed cytokine profile, providing the optimal MΦ stimulation that favored PS exposure on intracellular amastigotes. Maintenance of PS exposed on the parasite was dependent on low, but sustained, levels of nitric oxide and polyamine production. Amastigotes obtained from lymphopenic nude mice did not expose PS on their surface, and adoptive transfer of CD4+ T cells reversed this phenotype. In addition, histopathological analysis of mice treated with anti-PS antibodies showed increased inflammation and similarities to nude mouse lesions. Collectively, our data confirm the role of pathogenic CD4+ T cells for disease progression and point to PS as a critical parasite strategy to subvert host immune responses.
Collapse
Affiliation(s)
- Joao Luiz Mendes Wanderley
- Laboratório de Imunoparasitologia, Unidade de Pesquisa Integrada em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Macaé, Brazil
| | - Poliana Deolindo
- Laboratório de Biologia Molecular de Parasitas e Vetores, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Eric Carlsen
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Arieli Bernardo Portugal
- Laboratório de Imunoparasitologia, Unidade de Pesquisa Integrada em Produtos Bioativos e Biociências, Universidade Federal do Rio de Janeiro, Macaé, Brazil
| | - Renato Augusto DaMatta
- Laboratório de Biologia Celular e Tecidual, Universidade Estadual do Norte Fluminense, Campos dos Goytacazes, Brazil
| | - Marcello Andre Barcinski
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lynn Soong
- Department of Microbiology and Immunology, Center for Tropical Diseases, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, United States
| |
Collapse
|
22
|
Schiattarella GG, Altamirano F, Tong D, French KM, Villalobos E, Kim SY, Luo X, Jiang N, May HI, Wang ZV, Hill TM, Mammen PPA, Huang J, Lee DI, Hahn VS, Sharma K, Kass DA, Lavandero S, Gillette TG, Hill JA. Nitrosative stress drives heart failure with preserved ejection fraction. Nature 2019; 568:351-356. [PMID: 30971818 PMCID: PMC6635957 DOI: 10.1038/s41586-019-1100-z] [Citation(s) in RCA: 591] [Impact Index Per Article: 98.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 03/07/2019] [Indexed: 12/21/2022]
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a common syndrome with high morbidity and mortality for which there are no evidence-based therapies. Here we report that concomitant metabolic and hypertensive stress in mice-elicited by a combination of high-fat diet and inhibition of constitutive nitric oxide synthase using Nω-nitro-L-arginine methyl ester (L-NAME)-recapitulates the numerous systemic and cardiovascular features of HFpEF in humans. Expression of one of the unfolded protein response effectors, the spliced form of X-box-binding protein 1 (XBP1s), was reduced in the myocardium of our rodent model and in humans with HFpEF. Mechanistically, the decrease in XBP1s resulted from increased activity of inducible nitric oxide synthase (iNOS) and S-nitrosylation of the endonuclease inositol-requiring protein 1α (IRE1α), culminating in defective XBP1 splicing. Pharmacological or genetic suppression of iNOS, or cardiomyocyte-restricted overexpression of XBP1s, each ameliorated the HFpEF phenotype. We report that iNOS-driven dysregulation of the IRE1α-XBP1 pathway is a crucial mechanism of cardiomyocyte dysfunction in HFpEF.
Collapse
Affiliation(s)
- Gabriele G Schiattarella
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Francisco Altamirano
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dan Tong
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kristin M French
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elisa Villalobos
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Soo Young Kim
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiang Luo
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nan Jiang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Herman I May
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhao V Wang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Theodore M Hill
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pradeep P A Mammen
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jian Huang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dong I Lee
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Virginia S Hahn
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Kavita Sharma
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Sergio Lavandero
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical & Pharmaceutical Sciences and Faculty of Medicine, University of Chile, Santiago, Chile
- Center for Molecular Studies of the Cell (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Thomas G Gillette
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph A Hill
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
23
|
Hanoteau A, Newton JM, Krupar R, Huang C, Liu HC, Gaspero A, Gartrell RD, Saenger YM, Hart TD, Santegoets SJ, Laoui D, Spanos C, Parikh F, Jayaraman P, Zhang B, Van der Burg SH, Van Ginderachter JA, Melief CJM, Sikora AG. Tumor microenvironment modulation enhances immunologic benefit of chemoradiotherapy. J Immunother Cancer 2019; 7:10. [PMID: 30646957 PMCID: PMC6332704 DOI: 10.1186/s40425-018-0485-9] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 12/13/2018] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Chemoradiotherapy (CRT) remains one of the most common cancer treatment modalities, and recent data suggest that CRT is maximally effective when there is generation of an anti-tumoral immune response. However, CRT has also been shown to promote immunosuppressive mechanisms which must be blocked or reversed to maximize its immune stimulating effects. METHODS Therefore, using a preclinical model of human papillomavirus (HPV)-associated head and neck squamous cell carcinoma (HNSCC), we developed a clinically relevant therapy combining CRT and two existing immunomodulatory drugs: cyclophosphamide (CTX) and the small molecule inducible nitric oxide synthase (iNOS) inhibitor L-n6-(1-iminoethyl)-lysine (L-NIL). In this model, we treated the syngeneic HPV-HNSCC mEER tumor-bearing mice with fractionated (10 fractions of 3 Gy) tumor-directed radiation and weekly cisplatin administration. We compared the immune responses induced by CRT and those induced by combinatory treatment (CRT + CTX/L-NIL) with flow cytometry, quantitative multiplex immunofluorescence and by profiling immune-related gene expression changes. RESULTS We show that combination treatment favorably remodels the tumor myeloid immune microenvironment including an increase in anti-tumor immune cell types (inflammatory monocytes and M1-like macrophages) and a decrease in immunosuppressive granulocytic myeloid-derived suppressor cells (MDSCs). Intratumoral T cell infiltration and tumor antigen specificity of T cells were also improved, including a 31.8-fold increase in the CD8+ T cell/ regulatory T cell ratio and a significant increase in tumor antigen-specific CD8+ T cells compared to CRT alone. CTX/LNIL immunomodulation was also shown to significantly improve CRT efficacy, leading to rejection of 21% established tumors in a CD8-dependent manner. CONCLUSIONS Overall, these data show that modulation of the tumor immune microenvironment with CTX/L-NIL enhances susceptibility of treatment-refractory tumors to CRT. The combination of tumor immune microenvironment modulation with CRT constitutes a translationally relevant approach to enhance CRT efficacy through enhanced immune activation.
Collapse
Affiliation(s)
- Aurelie Hanoteau
- Department of Otolaryngology-Head and Neck surgery, Baylor College of Medicine, Houston, TX USA
| | - Jared M. Newton
- Department of Otolaryngology-Head and Neck surgery, Baylor College of Medicine, Houston, TX USA
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX USA
| | - Rosemarie Krupar
- Pathology of the University Hospital Schleswig-Holstein, Campus Luebeck and Research Center Borstel, Leibniz Lung Center, Lubeck and Borstel, Germany
| | - Chen Huang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX USA
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX USA
| | - Hsuan-Chen Liu
- Department of Otolaryngology-Head and Neck surgery, Baylor College of Medicine, Houston, TX USA
- Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX USA
| | - Angelina Gaspero
- Department of Otolaryngology-Head and Neck surgery, Baylor College of Medicine, Houston, TX USA
| | - Robyn D. Gartrell
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, Columbia University Irving Medical Center/New York Presbyterian, New York, USA
| | - Yvonne M. Saenger
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center/New York Presbyterian, New York, USA
| | - Thomas D. Hart
- Department of Medicine, Division of Hematology/Oncology, Columbia University Irving Medical Center/New York Presbyterian, New York, USA
| | - Saskia J. Santegoets
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Damya Laoui
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
| | - Chad Spanos
- Department of Surgery, University of South Dakota Sanford School of Medicine, Vermillion, SD USA
| | - Falguni Parikh
- Department of Otolaryngology-Head and Neck surgery, Baylor College of Medicine, Houston, TX USA
| | - Padmini Jayaraman
- Department of Otolaryngology-Head and Neck surgery, Baylor College of Medicine, Houston, TX USA
| | - Bing Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX USA
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX USA
| | - Sjoerd H. Van der Burg
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jo A. Van Ginderachter
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Laboratory of Myeloid Cell Immunology, VIB Center for Inflammation Research, Brussels, Belgium
| | | | - Andrew G. Sikora
- Department of Otolaryngology-Head and Neck surgery, Baylor College of Medicine, Houston, TX USA
- Department of Cell and Gene Therapy, Baylor College of Medicine, Houston, TX USA
| |
Collapse
|
24
|
Elkamhawy A, Hassan AHE, Paik S, Sup Lee Y, Lee HH, Shin JS, Lee KT, Roh EJ. EGFR inhibitors from cancer to inflammation: Discovery of 4-fluoro-N-(4-(3-(trifluoromethyl)phenoxy)pyrimidin-5-yl)benzamide as a novel anti-inflammatory EGFR inhibitor. Bioorg Chem 2019; 86:112-118. [PMID: 30685642 DOI: 10.1016/j.bioorg.2019.01.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 01/08/2019] [Accepted: 01/08/2019] [Indexed: 12/17/2022]
Abstract
EGFR inhibitors are well-known as anticancer agents. Quite differently, we report our effort to develop EGFR inhibitors as anti-inflammatory agents. Pyrimidinamide EGFR inhibitors eliciting low micromolar IC50 and the structurally close non-EGFR inhibitor urea analog were synthesized. Comparing their nitric oxide (NO) production inhibitory activity in peritoneal macrophages and RAW 246.7 macrophages indicated that their anti-inflammatory activity in peritoneal macrophages might be a sequence of EGFR inhibition. Further evaluations proved that compound 4d significantly and dose-dependently inhibits LPS-induced iNOS expression and IL-1β, IL-6, and TNF-α production via NF-κB inactivation in peritoneal macrophages. Compound 4d might serve as a lead compound for development of a novel class of anti-inflammatory EGFR inhibitors.
Collapse
Affiliation(s)
- Ahmed Elkamhawy
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Ahmed H E Hassan
- Department of Medicinal Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Medicinal Chemistry Laboratory, Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Sora Paik
- Department of Fundamental Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yong Sup Lee
- Medicinal Chemistry Laboratory, Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Fundamental Pharmaceutical Sciences, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hwi-Ho Lee
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ji-Sun Shin
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Kyung-Tae Lee
- Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Eun Joo Roh
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea.
| |
Collapse
|
25
|
Abstract
Nitric oxide (NO·) produced by mammalian cells exerts antimicrobial actions that result primarily from the modification of protein thiols (S-nitrosylation) and metal centers. A comprehensive approach was used to identify novel targets of NO· in Salmonella enterica serovar Typhimurium (S. Typhimurium). Newly identified targets include zinc metalloproteins required for DNA replication and repair (DnaG, PriA, and TopA), protein synthesis (AlaS and RpmE), and various metabolic activities (ClpX, GloB, MetE, PepA, and QueC). The cytotoxic actions of free zinc are mitigated by the ZntA and ZitB zinc efflux transporters, which are required for S. Typhimurium resistance to zinc overload and nitrosative stress in vitro Zinc efflux also ameliorates NO·-dependent zinc mobilization following internalization by activated macrophages and is required for virulence in NO·-producing mice, demonstrating that host-derived NO· causes zinc stress in intracellular bacteria.IMPORTANCE Nitric oxide (NO·) is produced by macrophages in response to inflammatory stimuli and restricts the growth of intracellular bacteria. Mechanisms of NO·-dependent antimicrobial actions are incompletely understood. Here, we show that zinc metalloproteins are important targets of NO· in Salmonella, including the DNA replication proteins DnaG and PriA, which were hypothesized to be NO· targets in earlier studies. Like iron, zinc is a cofactor for several essential proteins but is toxic at elevated concentrations. This study demonstrates that NO· mobilizes free zinc in Salmonella and that specific efflux transporters ameliorate the cytotoxic effects of free zinc during infection.
Collapse
|
26
|
Gupta S, Ahsan AU, Wani A, Khajuria V, Nazir LA, Sharma S, Bhagat A, Raj Sharma P, Bhardwaj S, Peerzada KJ, Ali Shah B, Ahmed Z. The amino analogue of β-boswellic acid efficiently attenuates the release of pro-inflammatory mediators than its parent compound through the suppression of NF-κB/IκBα signalling axis. Cytokine 2018; 107:93-104. [DOI: 10.1016/j.cyto.2017.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/01/2017] [Accepted: 12/03/2017] [Indexed: 11/29/2022]
|
27
|
CAR T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-1 blockade. Nat Med 2018; 24:731-738. [PMID: 29808005 DOI: 10.1038/s41591-018-0041-7] [Citation(s) in RCA: 955] [Impact Index Per Article: 136.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 03/14/2018] [Indexed: 01/06/2023]
Abstract
Chimeric antigen receptor (CAR) therapy targeting CD19 is an effective treatment for refractory B cell malignancies, especially acute lymphoblastic leukemia (ALL) 1 . Although a majority of patients will achieve a complete response following a single infusion of CD19-targeted CAR-modified T cells (CD19 CAR T cells)2-4, the broad applicability of this treatment is hampered by severe cytokine release syndrome (CRS), which is characterized by fever, hypotension and respiratory insufficiency associated with elevated serum cytokines, including interleukin-6 (IL-6)2,5. CRS usually occurs within days of T cell infusion at the peak of CAR T cell expansion. In ALL, it is most frequent and more severe in patients with high tumor burden2-4. CRS may respond to IL-6 receptor blockade but can require further treatment with high dose corticosteroids to curb potentially lethal severity2-9. Improved therapeutic and preventive treatments require a better understanding of CRS physiopathology, which has so far remained elusive. Here we report a murine model of CRS that develops within 2-3 d of CAR T cell infusion and that is potentially lethal and responsive to IL-6 receptor blockade. We show that its severity is mediated not by CAR T cell-derived cytokines, but by IL-6, IL-1 and nitric oxide (NO) produced by recipient macrophages, which enables new therapeutic interventions.
Collapse
|
28
|
Er H, Turkoz Y, Ozerol IH, Uzmez E. Effect of Nitric Oxide Synthase Inhibition in Experimental Pseudomonas keratitis in Rabbits. Eur J Ophthalmol 2018; 8:137-41. [PMID: 9793765 DOI: 10.1177/112067219800800303] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Purpose The aim of this study was to assess the efficacy of the nitric oxide synthase inhibitor L-NG-nitro-arginine-methyleser (L-NAME) in the treatment of experimental Pseudomonas keratitis. Methods Twelve young New Zealand white rabbits were given intrastromal injections of Pseudomonas aeruginosa strains. Twenty-four hours later, the rabbits were randomly divided into four groups: group 1 was treated with topical 0.3% ciprofloxacin drops and a single subconjunctival injection of L-NAME (150 mg/kg); group 2 received topical 0.3% ciprofloxacin drops alone; group 3 received a single subconjunctival injection of L-NAME alone; group 4, the control group, was treated with topical balanced salt solution (BSS) drops. One drop of either ciprofloxacin of BSS was applied at each treatment interval. Twenty-six hours after the start of treatment, the eyes were examined by slit lamp to assess inflammation. Aqueous humor specimens were collected from each eye to measure the nitric oxide concentration. Corneas were removed to count bacteria results. Results Slit lamp examination cell scores were significantly lower for groups 1 and 3 than groups 2 and 4 eyes (p = 0.002 to p = 0.004). No viable bacteria were detected in the corneas of groups 1 and 2. Group 3 corneas had significantly fewer bacteria (6.33 ± 0.42 × 103) than group 4 (5.94 ± 0.16 × 104) (p < 0.05). Nitric oxide levels in the aqueous humor were significantly lower for group 1 eyes than for groups 2, 3, or 4 (p = 0.02, p = 0.01, and p = 0.003, respectively). Conclusions We conclude that nitric oxide synthase inhibitors may be a useful adjunct but are not a replacement for traditional antibiotic drop therapy.
Collapse
Affiliation(s)
- H Er
- Department of Ophthalmology, Inönü University School of Medicine, Turgut Ozal Medical Center, Malatya, Turkey
| | | | | | | |
Collapse
|
29
|
Haraguchi Y, Mizoguchi Y, Ohgidani M, Imamura Y, Murakawa-Hirachi T, Nabeta H, Tateishi H, Kato TA, Monji A. Donepezil suppresses intracellular Ca 2+ mobilization through the PI3K pathway in rodent microglia. J Neuroinflammation 2017; 14:258. [PMID: 29273047 PMCID: PMC5741946 DOI: 10.1186/s12974-017-1033-0] [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: 07/28/2017] [Accepted: 12/11/2017] [Indexed: 12/17/2022] Open
Abstract
Background Microglia are resident innate immune cells which release many factors including proinflammatory cytokines or nitric oxide (NO) when they are activated in response to immunological stimuli. Pathophysiology of Alzheimer’s disease (AD) is related to the inflammatory responses mediated by microglia. Intracellular Ca2+ signaling is important for microglial functions such as release of NO and cytokines. In addition, alteration of intracellular Ca2+ signaling underlies the pathophysiology of AD, while it remains unclear how donepezil, an acetylcholinesterase inhibitor, affects intracellular Ca2+ mobilization in microglial cells. Methods We examined whether pretreatment with donepezil affects the intracellular Ca2+ mobilization using fura-2 imaging and tested the effects of donepezil on phagocytic activity by phagocytosis assay in rodent microglial cells. Results In this study, we observed that pretreatment with donepezil suppressed the TNFα-induced sustained intracellular Ca2+ elevation in both rat HAPI and mouse primary microglial cells. On the other hand, pretreatment with donepezil did not suppress the mRNA expression of both TNFR1 and TNFR2 in rodent microglia we used. Pretreatment with acetylcholine but not donepezil suppressed the TNFα-induced intracellular Ca2+ elevation through the nicotinic α7 receptors. In addition, sigma 1 receptors were not involved in the donepezil-induced suppression of the TNFα-mediated intracellular Ca2+ elevation. Pretreatment with donepezil suppressed the TNFα-induced intracellular Ca2+ elevation through the PI3K pathway in rodent microglial cells. Using DAF-2 imaging, we also found that pretreatment with donepezil suppressed the production of NO induced by TNFα treatment and the PI3K pathway could be important for the donepezil-induced suppression of NO production in rodent microglial cells. Finally, phagocytosis assay showed that pretreatment with donepezil promoted phagocytic activity of rodent microglial cells through the PI3K but not MAPK/ERK pathway. Conclusions These suggest that donepezil could directly modulate the microglial function through the PI3K pathway in the rodent brain, which might be important to understand the effect of donepezil in the brain. Electronic supplementary material The online version of this article (10.1186/s12974-017-1033-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yoshinori Haraguchi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Yoshito Mizoguchi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan.
| | - Masahiro Ohgidani
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshiomi Imamura
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Toru Murakawa-Hirachi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Hiromi Nabeta
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Hiroshi Tateishi
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| | - Takahiro A Kato
- Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Akira Monji
- Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga, 849-8501, Japan
| |
Collapse
|
30
|
Farag AK, Elkamhawy A, Londhe AM, Lee KT, Pae AN, Roh EJ. Novel LCK/FMS inhibitors based on phenoxypyrimidine scaffold as potential treatment for inflammatory disorders. Eur J Med Chem 2017; 141:657-675. [PMID: 29107425 DOI: 10.1016/j.ejmech.2017.10.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/19/2017] [Accepted: 10/02/2017] [Indexed: 01/24/2023]
Abstract
Tyrosine kinases including LCK and FMS are involved in inflammatory disorders as well as many types of cancer. Our team has designed and synthesized thirty novel pyrimidine based inhibitors targeting LCK, classified into four different series (amides, ureas, imines (Schiff base) and benzylamines). Twelve of them showed nanomolar IC50 values. Compound 7g showed excellent selectivity profile and was selectively potent over FMS kinase (IC50 value of 4.6 nM). Molecular docking study was performed to help us rationalize the obtained results and predict the possible binding mode for our compounds in both LCK and FMS. Based on the obtained biological assay data and modelling results, a detailed SAR study was discussed. As a further testing regarding the anti-inflammatory effect of the new compounds, in vitro cellular assay over RAW 264.7 macrophages was performed. Compound 7g exhibited excellent anti-inflammatory effect. Therefore, we report the design of novel phenoxypyrimidine derivatives as potent and selective LCK inhibitors and the discovery of 7g as potent and selective FMS/LCK dual inhibitor for the potential application in inflammatory disorders including rheumatoid arthritis (RA).
Collapse
Affiliation(s)
- Ahmed Karam Farag
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ahmed Elkamhawy
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Ashwini M Londhe
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Kyung-Tae Lee
- Department of Life and Nanopharmaceutical Science, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Ae Nim Pae
- Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea; Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Eun Joo Roh
- Chemical Kinomics Research Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science &Technology, KIST School, Korea University of Science and Technology, Seoul 02792, Republic of Korea.
| |
Collapse
|
31
|
Kiang JG. Exacerbation of Mild Hypoxia on Acute Radiation Syndrome and Subsequent Mortality. ADAPTIVE MEDICINE 2017; 9:28-33. [PMID: 34616568 PMCID: PMC8491646 DOI: 10.4247/am.2017.abg170] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mild hypoxia induced by 20% hemorrhage results in increases in few cytokine concentrations and sclerostin levels in blood, but shows no changes in bone formation, bone marrow cellularity, and gastrointestinal (GI) integrity and no systemic bacterial infection as well as no subsequent mortality. On the other hand, severe hypoxia induced by 40% hemorrhage causes significant increases in most cytokine concentrations, GI injury, lung injury, systemic bacterial infection, cellular ATP reduction and subsequent mortality. The severe hypoxia drastically damages GI and lung morphology, elevates cytokine concentrations in blood and increases inducible nitric oxide synthase (iNOS) expression in cells that is mediated by transcription factors NF-κB/NF-IL6, subsequently producing free radicals that disrupt mitochondria. ATP depletion, p53 phosphorylation, and caspase-3 activation are found, suggesting cell apoptosis. As a result, mortality occurs. However, when mild hypoxia follows ionizing radiation, the mild hypoxia significantly enhances radiation-induced mortality and acute radiation syndrome, including injury of bone marrow, GI, kidney, and lung. The synergism also occurs at the molecular level, resulting in alteration of microRNAs, amplification of iNOS expression, cytokine increases, sepsis, and ATP depletion. This is the first demonstration of synergistic effects between mild hypoxia and ionizing radiation.
Collapse
Affiliation(s)
- Juliann G Kiang
- Radiation Combined Injury Program, Armed Forces Radiobiology Research Institute Department of Pharmacology and Molecular Therapeutics, Department of Medicine Uniformed Services University of the Health Sciences, Bethesda, Maryland, U.S.A
| |
Collapse
|
32
|
Park KK, Reuben JS, Soliman KF. The Role of Inducible-Nitric Oxide in Cocaine-Induced Kindling. Exp Biol Med (Maywood) 2016; 226:185-90. [PMID: 11361036 DOI: 10.1177/153537020122600305] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Experimentally naive male Sprague Dawley rats (weighing 85–110 g) were used to examine the role of inducible nitric oxide synthase (iNOS) in cocaine-induced kindling. Repeated administration of cocaine (45 mg/kg, ip) to Sprague Dawley male rats for 7 consecutive days produced a progressive increase in the Convulsive responsiveness and death. Pretreatment with iNOS inhibitors, L-N6-(1-iminoethyl)lysine (NIL; 10 mg/kg, ip) and (–)-ePigalloocatechin gallate (EGCG; 10 mg/kg, ip) 30 min before cocaine (45 mg/kg, ip) administration for 7 days attenuated the development of cocaine kindling and blocked cocaine-induced death. Results of NMDA receptor binding assay in the hippocampus showed a significant increase in the affinity without changes in the density in animals treated with cocaine, but there were no changes in these parameters in the cortex. Pretreatment with NIL or EGCG prior to cocaine administration abolished the cocaine-induced effect in the NMDA receptor affinity in the hippocampus. These results suggest that iNOS induction followed by an increase of NMDA receptor affinity in the hippocampus after repeated exposure to cocaine may participate in the process of the development of cocaine kindling.
Collapse
Affiliation(s)
- K K Park
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee 32307, USA
| | | | | |
Collapse
|
33
|
Vielma AZ, León L, Fernández IC, González DR, Boric MP. Nitric Oxide Synthase 1 Modulates Basal and β-Adrenergic-Stimulated Contractility by Rapid and Reversible Redox-Dependent S-Nitrosylation of the Heart. PLoS One 2016; 11:e0160813. [PMID: 27529477 PMCID: PMC4986959 DOI: 10.1371/journal.pone.0160813] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 06/21/2016] [Indexed: 12/30/2022] Open
Abstract
S-nitrosylation of several Ca2+ regulating proteins in response to β-adrenergic stimulation was recently described in the heart; however the specific nitric oxide synthase (NOS) isoform and signaling pathways responsible for this modification have not been elucidated. NOS-1 activity increases inotropism, therefore, we tested whether β-adrenergic stimulation induces NOS-1-dependent S-nitrosylation of total proteins, the ryanodine receptor (RyR2), SERCA2 and the L-Type Ca2+ channel (LTCC). In the isolated rat heart, isoproterenol (10 nM, 3-min) increased S-nitrosylation of total cardiac proteins (+46±14%) and RyR2 (+146±77%), without affecting S-nitrosylation of SERCA2 and LTCC. Selective NOS-1 blockade with S-methyl-L-thiocitrulline (SMTC) and Nω-propyl-l-arginine decreased basal contractility and relaxation (−25–30%) and basal S-nitrosylation of total proteins (−25–60%), RyR2, SERCA2 and LTCC (−60–75%). NOS-1 inhibition reduced (−25–40%) the inotropic response and protein S-nitrosylation induced by isoproterenol, particularly that of RyR2 (−85±7%). Tempol, a superoxide scavenger, mimicked the effects of NOS-1 inhibition on inotropism and protein S-nitrosylation; whereas selective NOS-3 inhibitor L-N5-(1-Iminoethyl)ornithine had no effect. Inhibition of NOS-1 did not affect phospholamban phosphorylation, but reduced its oligomerization. Attenuation of contractility was abolished by PKA blockade and unaffected by guanylate cyclase inhibition. Additionally, in isolated mouse cardiomyocytes, NOS-1 inhibition or removal reduced the Ca2+-transient amplitude and sarcomere shortening induced by isoproterenol or by direct PKA activation. We conclude that 1) normal cardiac performance requires basal NOS-1 activity and S-nitrosylation of the calcium-cycling machinery; 2) β-adrenergic stimulation induces rapid and reversible NOS-1 dependent, PKA and ROS-dependent, S-nitrosylation of RyR2 and other proteins, accounting for about one third of its inotropic effect.
Collapse
Affiliation(s)
- Alejandra Z. Vielma
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, PO Box 114-D, Santiago, Chile
| | - Luisa León
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, PO Box 114-D, Santiago, Chile
| | - Ignacio C. Fernández
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, PO Box 114-D, Santiago, Chile
| | - Daniel R. González
- Departamento de Ciencias Básicas Biomédicas, Facultad de Ciencias de la Salud, Universidad de Talca, Av. Lircay S.N., Talca, Chile
| | - Mauricio P. Boric
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, PO Box 114-D, Santiago, Chile
- * E-mail:
| |
Collapse
|
34
|
Potential of Inducible Nitric Oxide Synthase as a Therapeutic Target for Allergen-Induced Airway Hyperresponsiveness: A Critical Connection to Nitric Oxide Levels and PARP Activity. Mediators Inflamm 2016; 2016:1984703. [PMID: 27524861 PMCID: PMC4971330 DOI: 10.1155/2016/1984703] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 05/11/2016] [Accepted: 05/22/2016] [Indexed: 12/20/2022] Open
Abstract
Although expression of inducible NO synthase (iNOS) in the lungs of asthmatics and associated nitrosative damage are established, iNOS failed as a therapeutic target for blocking airway hyperresponsiveness (AHR) and inflammation in asthmatics. This dichotomy calls for better strategies with which the enzyme is adequately targeted. Here, we confirm iNOS expression in the asthmatic lung with concomitant protein nitration and poly(ADP-ribose) polymerase (PARP) activation. We show, for the first time, that iNOS is highly expressed in peripheral blood mononuclear cells (PBMCs) of asthmatics with uncontrolled disease, which did not correspond to protein nitration. Selective iNOS inhibition with L-NIL protected against AHR upon acute, but not chronic, exposure to ovalbumin or house dust mite (HDM) in mice. Supplementation of NO by nitrite administration significantly blocked AHR in chronically HDM-exposed mice that were treated with L-NIL. Protection against chronic HDM exposure-induced AHR by olaparib-mediated PARP inhibition may be associated with the partial but not the complete blockade of iNOS expression. Indeed, L-NIL administration prevented olaparib-mediated protection against AHR in chronically HDM-exposed mice. Our study suggests that the amount of iNOS and NO are critical determinants in the modulation of AHR by selective iNOS inhibitors and renews the potential of iNOS as a therapeutic target for asthma.
Collapse
|
35
|
Wei W, Wu XW, Deng GG, Yang XW. Anti-inflammatory coumarins with short- and long-chain hydrophobic groups from roots of Angelica dahurica cv. Hangbaizhi. PHYTOCHEMISTRY 2016; 123:58-68. [PMID: 26775737 DOI: 10.1016/j.phytochem.2016.01.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 01/01/2016] [Accepted: 01/07/2016] [Indexed: 06/05/2023]
Abstract
The (1)H NMR-guided fractionation of a cyclohexane soluble portion of the 75% ethanolic extract of the roots of Angelica dahurica cv. Hangbaizhi led to the isolation of two coumarins, namely, 5-(3"-hydroxy-3"-methylbutyl)-8-hydroxyfuranocoumarin, and isobyakangelicin hydrate-3"-ethyl ether, and ten coumarins with short- or long-chain hydrophobic groups, namely, andafocoumarins A-J. Their structures were elucidated by extensive spectroscopic analyses. The absolute configurations of the C-2" secondary alcohols in ten of these compounds were deduced via the circular dichroism data of the in situ formed [Rh2(OCOCF3)4] complex, and oxidation reactions were utilized to determine location of the double bonds in the lipid chain of andafocoumarins H and I, respectively. The long-chain hydrophobic group of andafocoumarin J was determined by the method of chemical degradation and GC-MS analysis. It was the first time that coumarins with short- or long-chain hydrophobic groups in this plant had been comprehensively investigated. All isolates were assayed for their inhibitory effect against nitric oxide (NO) production in a lipopolysaccharide (LPS)-activated RAW264.7 macrophage cell line, among which andafocoumarins A and B exhibited a potent inhibition on LPS-activated NO production with IC50 values of 19.7 and 13.9 μM, respectively, indicating their stronger inhibitory activity than l-N(6)-(1-iminoethyl)-lysine (IC50=23.7 μM), a selective inhibitor of inducible nitric oxide synthase.
Collapse
Affiliation(s)
- Wei Wei
- State Key Laboratory of Natural and Biomimetic Drugs (Peking University), Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Peking University, Beijing 100191, PR China
| | - Xiu-Wen Wu
- State Key Laboratory of Natural and Biomimetic Drugs (Peking University), Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Peking University, Beijing 100191, PR China
| | - Gai-Gai Deng
- State Key Laboratory of Natural and Biomimetic Drugs (Peking University), Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Peking University, Beijing 100191, PR China
| | - Xiu-Wei Yang
- State Key Laboratory of Natural and Biomimetic Drugs (Peking University), Department of Natural Medicines, School of Pharmaceutical Sciences, Peking University Health Science Center, Peking University, Beijing 100191, PR China.
| |
Collapse
|
36
|
E1 of α-ketoglutarate dehydrogenase defends Mycobacterium tuberculosis against glutamate anaplerosis and nitroxidative stress. Proc Natl Acad Sci U S A 2015; 112:E5834-43. [PMID: 26430237 DOI: 10.1073/pnas.1510932112] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Enzymes of central carbon metabolism (CCM) in Mycobacterium tuberculosis (Mtb) make an important contribution to the pathogen's virulence. Evidence is emerging that some of these enzymes are not simply playing the metabolic roles for which they are annotated, but can protect the pathogen via additional functions. Here, we found that deficiency of 2-hydroxy-3-oxoadipate synthase (HOAS), the E1 component of the α-ketoglutarate (α-KG) dehydrogenase complex (KDHC), did not lead to general metabolic perturbation or growth impairment of Mtb, but only to the specific inability to cope with glutamate anaplerosis and nitroxidative stress. In the former role, HOAS acts to prevent accumulation of aldehydes, including growth-inhibitory succinate semialdehyde (SSA). In the latter role, HOAS can participate in an alternative four-component peroxidase system, HOAS/dihydrolipoyl acetyl transferase (DlaT)/alkylhydroperoxide reductase colorless subunit gene (ahpC)-neighboring subunit (AhpD)/AhpC, using α-KG as a previously undescribed source of electrons for reductase action. Thus, instead of a canonical role in CCM, the E1 component of Mtb's KDHC serves key roles in situational defense that contribute to its requirement for virulence in the host. We also show that pyruvate decarboxylase (AceE), the E1 component of pyruvate dehydrogenase (PDHC), can participate in AceE/DlaT/AhpD/AhpC, using pyruvate as a source of electrons for reductase action. Identification of these systems leads us to suggest that Mtb can recruit components of its CCM for reactive nitrogen defense using central carbon metabolites.
Collapse
|
37
|
Ligand activation of cannabinoid receptors attenuates hypertrophy of neonatal rat cardiomyocytes. J Cardiovasc Pharmacol 2015; 64:420-30. [PMID: 24979612 DOI: 10.1097/fjc.0000000000000134] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
: Endocannabinoids are bioactive amides, esters, and ethers of long-chain polyunsaturated fatty acids. Evidence suggests that activation of the endocannabinoid pathway offers cardioprotection against myocardial ischemia, arrhythmias, and endothelial dysfunction of coronary arteries. As cardiac hypertrophy is a convergence point of risk factors for heart failure, we determined a role for endocannabinoids in attenuating endothelin-1-induced hypertrophy and probed the signaling pathways involved. The cannabinoid receptor ligand anandamide and its metabolically stable analog, R-methanandamide, suppressed hypertrophic indicators including cardiomyocyte enlargement and fetal gene activation (ie, the brain natriuretic peptide gene) elicited by endothelin-1 in isolated neonatal rat ventricular myocytes. The ability of R-methanandamide to suppress myocyte enlargement and fetal gene activation was mediated by CB2 and CB1 receptors, respectively. Accordingly, a CB2-selective agonist, JWH-133, prevented only myocyte enlargement but not brain natriuretic peptide gene activation. A CB1/CB2 dual agonist with limited brain penetration, CB-13, inhibited both hypertrophic indicators. CB-13 activated AMP-activated protein kinase (AMPK) and, in an AMPK-dependent manner, endothelial nitric oxide synthase (eNOS). Disruption of AMPK signaling, using compound C or short hairpinRNA knockdown, and eNOS inhibition using L-NIO abolished the antihypertrophic actions of CB-13. In conclusion, CB-13 inhibits cardiomyocyte hypertrophy through AMPK-eNOS signaling and may represent a novel therapeutic approach to cardioprotection.
Collapse
|
38
|
Tang W, Li H, Poulos TL, Silverman RB. Mechanistic studies of inactivation of inducible nitric oxide synthase by amidines. Biochemistry 2015; 54:2530-8. [PMID: 25811913 DOI: 10.1021/acs.biochem.5b00135] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nitric oxide synthase (NOS) catalyzes the conversion of L-arginine to L-citrulline and nitric oxide. N(5)-(1-Iminoethyl)-L-ornithine (L-NIO), an amidine-containing molecule, is a natural product known to be an inactivator of inducible NOS (iNOS). Because of the presence of the amidine methyl group in place of the guanidine amino group of substrate L-arginine, the active site heme peroxy intermediate sometimes cannot be protonated, thereby preventing its conversion to the heme oxo intermediate; instead, a heme oxygenase-type mechanism occurs, leading to conversion of the heme to biliverdin. This might be a new and general inactivation mechanism for heme-containing enzymes. In the studies described here, we attempted to provide support for amidines as substrates and inactivators of iNOS by the design and synthesis of amidine analogues of L-NIO having groups other than the amidine methyl group. No nitric oxide- or enzyme-catalyzed products could be detected by incubation of these amidines with iNOS. Although none of the L-NIO analogues acted as substrates, they all inhibited iNOS; increased inhibitory potency correlated with decreased substituent size. Computer modeling and molecular dynamics simulations were run on 10 and 11 to rationalize why these compounds do not act as substrates. Unlike the methyl amidine (L-NIO), the other alkyl groups block binding of O2 at the heme iron. Compounds 8, 9, and 11 were inactivators; however, no heme was lost, and no biliverdin was formed. No kinetic isotope effect on inactivation was observed with perdeuterated ethyl 8. A small amount of dimer disruption occurred with these inactivators, although the amount would not account for complete enzyme inactivation. The L-NIO analogues inactivate iNOS by a yet unknown mechanism; however, it is different from that of L-NIO, and the inactivation mechanism previously reported for L-NIO appears to be unique to methyl amidines.
Collapse
Affiliation(s)
- Wei Tang
- †Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, and Center for Molecular Innovation and Drug Discovery, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
| | - Huiying Li
- ‡Departments of Molecular Biology and Biochemistry, Chemistry, and Pharmaceutical Sciences, University of California, Irvine, California 92697-3900, United States
| | - Thomas L Poulos
- ‡Departments of Molecular Biology and Biochemistry, Chemistry, and Pharmaceutical Sciences, University of California, Irvine, California 92697-3900, United States
| | - Richard B Silverman
- †Department of Chemistry, Department of Molecular Biosciences, Chemistry of Life Processes Institute, and Center for Molecular Innovation and Drug Discovery, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208-3113, United States
| |
Collapse
|
39
|
Devi KSP, Das J, Kumari K, Singh P, Behera B, Maiti TK. AMPK-mediated crosstalk of heteroglycan-induced reactive species and autophagic cascade in RAW 264.7 cells. RSC Adv 2015. [DOI: 10.1039/c5ra05127d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AMPK mediates the crosstalk among heteroglycan-induced autophagy and reactive species in RAW 264.7 cells.
Collapse
Affiliation(s)
- K. Sanjana P. Devi
- Department of Biotechnology
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| | - Joyjyoti Das
- Department of Biotechnology
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| | - Kalpana Kumari
- Department of Biotechnology
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| | - Prashant Singh
- Department of Biotechnology
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| | - Birendra Behera
- Department of Biotechnology
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| | - T. K. Maiti
- Department of Biotechnology
- Indian Institute of Technology Kharagpur
- Kharagpur 721302
- India
| |
Collapse
|
40
|
Shinozaki S, Chang K, Sakai M, Shimizu N, Yamada M, Tanaka T, Nakazawa H, Ichinose F, Yamada Y, Ishigami A, Ito H, Ouchi Y, Starr ME, Saito H, Shimokado K, Stamler JS, Kaneki M. Inflammatory stimuli induce inhibitory S-nitrosylation of the deacetylase SIRT1 to increase acetylation and activation of p53 and p65. Sci Signal 2014; 7:ra106. [PMID: 25389371 DOI: 10.1126/scisignal.2005375] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inflammation increases the abundance of inducible nitric oxide synthase (iNOS), leading to enhanced production of nitric oxide (NO), which can modify proteins by S-nitrosylation. Enhanced NO production increases the activities of the transcription factors p53 and nuclear factor κB (NF-κB) in several models of disease-associated inflammation. S-nitrosylation inhibits the activity of the protein deacetylase SIRT1. SIRT1 limits apoptosis and inflammation by deacetylating p53 and p65 (also known as RelA), a subunit of NF-κB. We showed in multiple cultured mammalian cell lines that NO donors or inflammatory stimuli induced S-nitrosylation of SIRT1 within CXXC motifs, which inhibited SIRT1 by disrupting its ability to bind zinc. Inhibition of SIRT1 reduced deacetylation and promoted activation of p53 and p65, leading to apoptosis and increased expression of proinflammatory genes. In rodent models of systemic inflammation, Parkinson's disease, or aging-related muscular atrophy, S-nitrosylation of SIRT1 correlated with increased acetylation of p53 and p65 and activation of p53 and NF-κB target genes, suggesting that S-nitrosylation of SIRT1 may represent a proinflammatory switch common to many diseases and aging.
Collapse
Affiliation(s)
- Shohei Shinozaki
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA. Department of Geriatrics and Vascular Medicine, Tokyo Medical and Dental University Graduate School, Tokyo 113-8519, Japan
| | - Kyungho Chang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA. Department of Anesthesiology and Pain Relief Center, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Michihiro Sakai
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA
| | - Nobuyuki Shimizu
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA
| | - Marina Yamada
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA
| | - Tomokazu Tanaka
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA
| | - Harumasa Nakazawa
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA
| | - Fumito Ichinose
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA
| | - Yoshitsugu Yamada
- Department of Anesthesiology and Pain Relief Center, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan
| | - Akihito Ishigami
- Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan
| | - Hideki Ito
- Tokyo Metropolitan Institute of Gerontology, Tokyo 173-0015, Japan
| | - Yasuyoshi Ouchi
- Department of Geriatric Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655, Japan. Federation of National Public Service Personnel Mutual Aid Associations Toranomon Hospital, Tokyo 105-0001, Japan
| | - Marlene E Starr
- Department of Surgery, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Hiroshi Saito
- Department of Surgery, University of Kentucky College of Medicine, Lexington, KY 40536, USA
| | - Kentaro Shimokado
- Department of Geriatrics and Vascular Medicine, Tokyo Medical and Dental University Graduate School, Tokyo 113-8519, Japan
| | - Jonathan S Stamler
- Institute for Transformative Molecular Medicine and Harrington Discovery Institute, Case Western Reserve University and University Hospital, Cleveland, OH 44106, USA
| | - Masao Kaneki
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Shriners Hospitals for Children, Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
41
|
Mao Y, Poschke I, Kiessling R. Tumour-induced immune suppression: role of inflammatory mediators released by myelomonocytic cells. J Intern Med 2014; 276:154-70. [PMID: 24597954 DOI: 10.1111/joim.12229] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumour-induced immune dysfunction is a serious challenge to immunotherapy for cancer, and intact adaptive and innate cellular immunity is key to its success. Myelomonocytic cells have a central role in this immune suppression, and tumour-associated macrophages, eosinophils, neutrophils and myeloid-derived suppressor cells have all been shown to be of major importance. These myelomonocytic cells secrete a broad repertoire of inflammatory mediators providing them with powerful tools to inhibit tumour-reactive T cells and natural killer cells; free oxygen radicals including reactive oxygen species and NO, arginase, indoleamine 2,3-dioxygenase, prostaglandins, the pro-inflammatory heterodimer S100A8/9 and cytokines, such as granulocyte-macrophage colony-stimulating factor and transforming growth factor-β, have proven particularly potent in suppressing antitumour cellular immunity. Determining which of these factors prevail in individual cancer patients and designing methods aimed at neutralization or inhibition of their effects on target tissues have the potential to greatly enhance the clinical efficacy of immunotherapy.
Collapse
Affiliation(s)
- Y Mao
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
42
|
Mizoguchi Y, Kato TA, Seki Y, Ohgidani M, Sagata N, Horikawa H, Yamauchi Y, Sato-Kasai M, Hayakawa K, Inoue R, Kanba S, Monji A. Brain-derived neurotrophic factor (BDNF) induces sustained intracellular Ca2+ elevation through the up-regulation of surface transient receptor potential 3 (TRPC3) channels in rodent microglia. J Biol Chem 2014; 289:18549-55. [PMID: 24811179 DOI: 10.1074/jbc.m114.555334] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Microglia are immune cells that release factors, including proinflammatory cytokines, nitric oxide (NO), and neurotrophins, following activation after disturbance in the brain. Elevation of intracellular Ca(2+) concentration ([Ca(2+)]i) is important for microglial functions such as the release of cytokines and NO from activated microglia. There is increasing evidence suggesting that pathophysiology of neuropsychiatric disorders is related to the inflammatory responses mediated by microglia. Brain-derived neurotrophic factor (BDNF) is a neurotrophin well known for its roles in the activation of microglia as well as in pathophysiology and/or treatment of neuropsychiatric disorders. In this study, we sought to examine the underlying mechanism of BDNF-induced sustained increase in [Ca(2+)]i in rodent microglial cells. We observed that canonical transient receptor potential 3 (TRPC3) channels contribute to the maintenance of BDNF-induced sustained intracellular Ca(2+) elevation. Immunocytochemical technique and flow cytometry also revealed that BDNF rapidly up-regulated the surface expression of TRPC3 channels in rodent microglial cells. In addition, pretreatment with BDNF suppressed the production of NO induced by tumor necrosis factor α (TNFα), which was prevented by co-adiministration of a selective TRPC3 inhibitor. These suggest that BDNF induces sustained intracellular Ca(2+) elevation through the up-regulation of surface TRPC3 channels and TRPC3 channels could be important for the BDNF-induced suppression of the NO production in activated microglia. We show that TRPC3 channels could also play important roles in microglial functions, which might be important for the regulation of inflammatory responses and may also be involved in the pathophysiology and/or the treatment of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yoshito Mizoguchi
- From the Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan,
| | - Takahiro A Kato
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, the Innovation Center for Medical Redox Navigation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and
| | - Yoshihiro Seki
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masahiro Ohgidani
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, the Innovation Center for Medical Redox Navigation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and
| | - Noriaki Sagata
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, the Innovation Center for Medical Redox Navigation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan, and
| | - Hideki Horikawa
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Yusuke Yamauchi
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Mina Sato-Kasai
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Kohei Hayakawa
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Ryuji Inoue
- the Department of Physiology, Fukuoka University School of Medicine, 7-45-1 Nanakuma, Jyonan-ku, Fukuoka 812-0180, Japan
| | - Shigenobu Kanba
- the Department of Neuropsychiatry, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Akira Monji
- From the Department of Psychiatry, Faculty of Medicine, Saga University, 5-1-1 Nabeshima, Saga 849-8501, Japan
| |
Collapse
|
43
|
Zhong HJ, Liu LJ, Chong CM, Lu L, Wang M, Chan DSH, Chan PWH, Lee SMY, Ma DL, Leung CH. Discovery of a natural product-like iNOS inhibitor by molecular docking with potential neuroprotective effects in vivo. PLoS One 2014; 9:e92905. [PMID: 24690920 PMCID: PMC3972188 DOI: 10.1371/journal.pone.0092905] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Accepted: 02/27/2014] [Indexed: 12/11/2022] Open
Abstract
In this study, we applied structure-based virtual screening techniques to identify natural product or natural product-like inhibitors of iNOS. The iNOS inhibitory activity of the hit compounds was characterized using cellular assays and an in vivo zebrafish larvae model. The natural product-like compound 1 inhibited NO production in LPS-stimulated Raw264.7 macrophages, without exerting cytotoxic effects on the cells. Significantly, compound 1 was able to reverse MPTP-induced locomotion deficiency and neurotoxicity in an in vivo zebrafish larval model. Hence, compound 1 could be considered as a scaffold for the further development of iNOS inhibitors for potential anti-inflammatory or anti-neurodegenerative applications.
Collapse
Affiliation(s)
- Hai-Jing Zhong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Li-Juan Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Cheong-Meng Chong
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Lihua Lu
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Modi Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Daniel Shiu-Hin Chan
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Philip Wai Hong Chan
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, Singapore, Singapore
| | - Simon Ming-Yuen Lee
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Dik-Lung Ma
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tong, Hong Kong, China
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
44
|
Nitric oxide synthase inhibitors prevent the growth-inhibiting effects of quinpirole. Optom Vis Sci 2014; 90:1167-75. [PMID: 24061155 DOI: 10.1097/opx.0000000000000041] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Both dopamine and nitric oxide (NO) have been implicated in the signal cascade mediating ocular growth inhibition. If both are part of the same pathway, which precedes the other? We tested the hypothesis that dopamine acts upstream of NO, by using two NOS inhibitors in combination with the dopamine agonist quinpirole, and measured the effects on ocular growth rate. METHODS Chicks wore -10 D lenses or diffusers (FD) for 4 days starting at age 13 days. Experimental eyes received daily 20 μL injections of the following: quinpirole-lens: n = 12, FD: n = 20; n-ω-propyl-L-arginine (NPA)-lens: n = 6, FD: n = 4; quinpirole + NPA-lens: n = 17, FD: n = 19; and quinpirole + L-NIO-lens: n = 12, FD: n = 12. Saline injections were done as controls. High-frequency ultrasonography was done at the start, and on day 5, prior to injections and 3 hours later. Refractions were measured on day 5. RESULTS As expected, quinpirole prevented the development of axial myopia in both paradigms. When quinpirole was combined with either NOS inhibitor, however, eyes became myopic compared to quinpirole (FD: NPA: -5.9 D vs. -3.4 D; L-NIO: -5.8 D vs. -3.4 D; lens: NPA: -3.5 D vs. -0.4 D; p < 0.05 for all; L-NIO was not significant). This was the result of a disinhibition of vitreous chamber growth versus quinpirole (FD: NPA: 401 vs. 275 μm/4 d; L-NIO: 440 vs. 275 μm/4 d; LENS: NPA: 407 vs. 253µm/4 d; L-NIO: 403 vs. 253 μm/4 d; p < 0.05). Only NPA prevented the quinpirole-induced choroidal thickening in lens-wearing eyes (0 vs. 31 μm/3 h; p < 0.05). Choroidal thickening was not inhibited by either drug in FD eyes. CONCLUSIONS Dopamine acts upstream of NO and the choroidal response in the signal cascade mediating ocular growth inhibition in both form deprivation and negative lens wear. That neither NOS inhibitor inhibits choroidal thickening in FD eyes suggests that the choroidal mechanisms differ in the two paradigms.
Collapse
|
45
|
Nguyen QPB, Kim BM, Song MS, Yoon KH, Chai KY. Inhibitory Effects of N-Amido-3,3-difluoropyrrolidin-2-ones on LPS-induced Nitric Oxide Production in RAW 264.7 Macrophages. B KOREAN CHEM SOC 2014. [DOI: 10.5012/bkcs.2014.35.1.313] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
46
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: enzymes. Br J Pharmacol 2013; 170:1797-867. [PMID: 24528243 PMCID: PMC3892293 DOI: 10.1111/bph.12451] [Citation(s) in RCA: 415] [Impact Index Per Article: 34.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Enzymes are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, nuclear hormone receptors, catalytic receptors and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
47
|
Yang Z, Misner B, Ji H, Poulos TL, Silverman RB, Meyskens FL, Yang S. Targeting nitric oxide signaling with nNOS inhibitors as a novel strategy for the therapy and prevention of human melanoma. Antioxid Redox Signal 2013; 19. [PMID: 23199242 PMCID: PMC3704054 DOI: 10.1089/ars.2012.4563] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
AIMS Our previous studies have shown that nitric oxide (NO) plays an important role in increasing the invasion and proliferation of human melanoma cells, suggesting that targeting NO signaling may facilitate therapy and prevention. Neuronal nitric oxide synthase (nNOS) is present in melanocytes, a cell type that originates from the neural crest. The aims of this study were to determine the role of nNOS in melanoma progression and the potential antitumor effects of novel synthesized nNOS inhibitors. RESULTS In vitro studies demonstrated abundant expression of nNOS in melanoma compared to melanocytes, which was inducible by ultraviolet radiation and was associated with increased NO generation. nNOS was also detected in melanoma biopsies that increased with disease stage. Knockdown of nNOS in melanoma cells diminished L-arginine-induced NO production; the metastatic capacity was also reduced as well as the levels of MMP-1, Bcl-2, JunD, and APE/Ref-1. Similar inhibition of NO and invasion potential was observed utilizing novel, highly selective nNOS inhibitors. In three-dimensional human skin reconstructs, the nNOS inhibitor cpd8 effectively reversed the melanoma overgrowth stimulated by NO stress. INNOVATION Our work lays the foundation for development of clinical "drug-like" nNOS inhibitors as a new and promising strategy for the chemoprevention of early melanoma progression and the inhibition of secondary melanoma in high-risk individuals. CONCLUSION Based on our observations, we propose that nNOS in melanoma results in constitutive overproduction of NO, which stimulates proliferation and increases invasion potential, leading to subsequent development of metastases.
Collapse
Affiliation(s)
- Zhen Yang
- Chao Family Comprehensive Cancer Center, University of California Irvine, Irvine, California, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Deshpande SR, Satyanarayana K, Rao MNA, Pai KV. Nitric oxide modulators: an emerging class of medicinal agents. Indian J Pharm Sci 2013; 74:487-97. [PMID: 23798773 PMCID: PMC3687917 DOI: 10.4103/0250-474x.110572] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 11/10/2012] [Accepted: 11/15/2012] [Indexed: 12/19/2022] Open
Abstract
Nitric oxide, a unique messenger in biological system, is ubiquitously present virtually in all tissues revealing its versatile nature of being involved in diverse physiological functions such as vascular tone, inhibition of platelet aggregation, cell adhesion, neurotransmission and enzyme and immune regulation. The tremendous advancements made in the past few decades in this area suggests that the nitric oxide modulation either by its exogenous release through nitric oxide donors or inhibition of its synthesis by nitric oxide synthase inhibitors in physiological milieu may provide newer clinical strategies for the treatment of some diseases. In this review, an attempt is made to document and understand the biological chemistry of different classes of nitric oxide modulators that would prove to be a fruitful area in the years to come.
Collapse
Affiliation(s)
- S R Deshpande
- Department of Medicinal and Pharmaceutical Chemistry, HSK College of Pharmacy, Bagalkote-587 101, India
| | | | | | | |
Collapse
|
49
|
Nitric oxide regulates cardiac intracellular Na⁺ and Ca²⁺ by modulating Na/K ATPase via PKCε and phospholemman-dependent mechanism. J Mol Cell Cardiol 2013; 61:164-71. [PMID: 23612119 PMCID: PMC3981027 DOI: 10.1016/j.yjmcc.2013.04.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Revised: 04/11/2013] [Accepted: 04/12/2013] [Indexed: 12/14/2022]
Abstract
In the heart, Na/K-ATPase regulates intracellular Na+ and Ca2 + (via NCX), thereby preventing Na+ and Ca2 + overload and arrhythmias. Here, we test the hypothesis that nitric oxide (NO) regulates cardiac intracellular Na+ and Ca2 + and investigate mechanisms and physiological consequences involved. Effects of both exogenous NO (via NO-donors) and endogenously synthesized NO (via field-stimulation of ventricular myocytes) were assessed in this study. Field stimulation of rat ventricular myocytes significantly increased endogenous NO (18 ± 2 μM), PKCε activation (82 ± 12%), phospholemman phosphorylation (at Ser-63 and Ser-68) and Na/K-ATPase activity (measured by DAF-FM dye, western-blotting and biochemical assay, respectively; p < 0.05, n = 6) and all were abolished by Ca2 +-chelation (EGTA 10 mM) or NOS inhibition l-NAME (1 mM). Exogenously added NO (spermine-NONO-ate) stimulated Na/K-ATPase (EC50 = 3.8 μM; n = 6/grp), via decrease in Km, in PLMWT but not PLMKO or PLM3SA myocytes (where phospholemman cannot be phosphorylated) as measured by whole-cell perforated-patch clamp. Field-stimulation with l-NAME or PKC-inhibitor (2 μM Bis) resulted in elevated intracellular Na+ (22 ± 1.5 and 24 ± 2 respectively, vs. 14 ± 0.6 mM in controls) in SBFI-AM-loaded rat myocytes. Arrhythmia incidence was significantly increased in rat hearts paced in the presence of l-NAME (and this was reversed by l-arginine), as well as in PLM3SA mouse hearts but not PLMWT and PLMKO. We provide physiological and biochemical evidence for a novel regulatory pathway whereby NO activates Na/K-ATPase via phospholemman phosphorylation and thereby limits Na+ and Ca2 + overload and arrhythmias. This article is part of a Special Issue entitled “Na+ Regulation in Cardiac Myocytes”.
We tested whether nitric oxide regulates intracellular Na+ and Ca2 + in the heart. Nitric oxide increased Na/K ATPase activity via PKCε-induced phospholemman phosphorylation. Inhibiting nitric oxide pathway resulted in Na+ and Ca2 + overload and contributed to arrhythmia development in the heart.
Collapse
|
50
|
Park JE, Abrams MJ, Efron PA, Barbul A. Excessive nitric oxide impairs wound collagen accumulation. J Surg Res 2012; 183:487-92. [PMID: 23290597 DOI: 10.1016/j.jss.2012.11.056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 10/19/2012] [Accepted: 11/30/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND Nitric oxide (NO) plays a major regulatory role in wound collagen synthesis. We hypothesized that this regulatory role is tightly controlled by the levels of NO in the wound environment and that supranormal wound NO generation impairs wound collagen accumulation. MATERIALS AND METHODS We used the model of turpentine-induced granuloma in male Sprague-Dawley rats as a sterile inflammatory stimulus generating large amounts of NO. In this environment, NO generation increased by 260%, whereas collagen deposition was significantly reduced by 38.5% (729.7 ± 81.5 versus 449.4 ± 76.3 μg hydroxyproline/100 mg sponge, P<0.05). Inhibition of NO synthase activity using 300 mM L-N6-(1-iminoethyl)-lysine, a highly potent and selective inhibitor of inducible NO synthase, significantly reduced NO elevation by 43.3% and increased wound collagen deposition by 37.3% (P<0.05). These effects occurred without any anti-inflammatory effects of L-N6-(1-iminoethyl)-lysine as assessed by the white blood cell counts and levels of interleukins 1 and 6. CONCLUSIONS The data show that high levels of NO within the wound environment significantly reduce wound collagen deposition. Inhibition of NO generation restores collagen levels to normal levels. The regulatory effects of NO on wound collagen appear to be highly correlated with the amount of NO generated.
Collapse
Affiliation(s)
- Julie E Park
- Department of Surgery, Sinai Hospital of Baltimore, Baltimore, Maryland, USA
| | | | | | | |
Collapse
|