1
|
Lu Y, Gu Y, Chan ASL, Yung Y, Wong YH. Activation of Bradykinin B 2 Receptors in Astrocytes Stimulates the Release of Leukemia Inhibitory Factor for Autocrine and Paracrine Signaling. Int J Mol Sci 2024; 25:13079. [PMID: 39684791 DOI: 10.3390/ijms252313079] [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: 09/30/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Abstract
Communications between different cell types within a tissue are often critical for the proper functioning of an organ. In the central nervous system, interactions among neurons and glial cells are known to modulate neurotransmission, energy metabolism, extracellular ion homeostasis, and neuroprotection. Here we showed that bradykinin, a proinflammatory neuropeptide, can be detected by astrocytes, resulting in the secretion of cytokines that act on neurons. In astrocytic cell lines and primary astrocytes, bradykinin and several other ligands acting on Gq-coupled receptors stimulated Ca2+ mobilization, which subsequently led to the release of leukemia inhibitory factor (LIF) and interleukin-6 (IL-6). The bradykinin B2 receptor antagonist, HOE-140, effectively blocked the ability of bradykinin to mobilize Ca2+ and stimulate mitogen-activated protein kinases (MAPKs) in astrocytes. Interestingly, incubation of neuronal cell lines and primary cortical neurons with conditioned media from bradykinin-treated astrocytes resulted in the activation of STAT3, a key component downstream of LIF and IL-6 receptors. LIF was apparently the major active factor in the conditioned media as the STAT3 response was almost completely neutralized by an anti-LIF antiserum. The presence of kininogen and kallikrein transcripts in neuronal cells but not in astrocytic cells indicates that neurons can produce bradykinin. Correspondingly, conditioned media from neuronal cells stimulated MAPKs in astrocytes in a HOE-140-sensitive manner. These studies demonstrate that paracrine signaling between neurons and astrocytes may involve ligands of Gq-coupled receptors and cytokines such as LIF.
Collapse
Affiliation(s)
- Ying Lu
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- School of Public Health, Nantong University, Nantong City 226019, China
| | - Yishan Gu
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Anthony S L Chan
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ying Yung
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Yung H Wong
- Division of Life Science and the Biotechnology Research Institute, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- State Key Laboratory of Molecular Neuroscience, and the Molecular Neuroscience Center, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, InnoHK, Hong Kong, China
| |
Collapse
|
2
|
Breault É, Desgagné M, Neve JD, Côté J, Barlow TMA, Ballet S, Sarret P. Multitarget ligands that comprise opioid/nonopioid pharmacophores for pain management: Current state of the science. Pharmacol Res 2024; 209:107408. [PMID: 39307212 DOI: 10.1016/j.phrs.2024.107408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 10/05/2024]
Abstract
Chronic pain, which affects more than one-third of the world's population, represents one of the greatest medical challenges of the 21st century, yet its effective management remains sub-optimal. The 'gold standard' for the treatment of moderate to severe pain consists of opioid ligands, such as morphine and fentanyl, that target the µ-opioid receptor (MOP). Paradoxically, these opioids also cause serious side effects, including constipation, respiratory depression, tolerance, and addiction. In addition, the development of opioid-use disorders, such as opioid diversion, misuse, and abuse, has led to the current opioid crisis, with dramatic increases in addiction, overdoses, and ultimately deaths. As pain is a complex, multidimensional experience involving a variety of pathways and mediators, dual or multitarget ligands that can bind to more than one receptor and exert complementary analgesic effects, represent a promising avenue for pain relief. Indeed, unlike monomodal therapeutic approaches, the modulation of several endogenous nociceptive systems can often result in an additive or even synergistic effect, thereby improving the analgesic-to-side-effect ratio. Here, we provide a comprehensive overview of research efforts towards the development of dual- or multi-targeting opioid/nonopioid hybrid ligands for effective and safer pain management. We reflect on the underpinning discovery rationale by discussing the design, medicinal chemistry, and in vivo pharmacological effects of multitarget antinociceptive compounds.
Collapse
Affiliation(s)
- Émile Breault
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Michael Desgagné
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Jolien De Neve
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, Brussels 1050, Belgium
| | - Jérôme Côté
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e avenue Nord, Sherbrooke, QC J1H 5N4, Canada
| | - Thomas M A Barlow
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, Brussels 1050, Belgium
| | - Steven Ballet
- Research Group of Organic Chemistry, Departments of Chemistry and Bioengineering Sciences, Vrije Universiteit Brussel, Pleinlaan 2, Brussels 1050, Belgium
| | - Philippe Sarret
- Institut de Pharmacologie de Sherbrooke, Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, 3001, 12e avenue Nord, Sherbrooke, QC J1H 5N4, Canada.
| |
Collapse
|
3
|
Li P, Lu H, Shi X, Yan J, Zhou L, Yang J, Wang B, Zhao Y, Liu L, Zhu Y, Xu L, Yang X, Su X, Yang Y, Zhang T, Guo L, Liu X. Protective effects of human urinary kallidinogenase against corticospinal tract damage in acute ischemic stroke patients. Neuroreport 2024; 35:431-438. [PMID: 38526971 DOI: 10.1097/wnr.0000000000002028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
This study aimed to assess the effects of human urinary kallidinogenase (HUK) on motor function outcome and corticospinal tract recovery in patients with acute ischemic stroke (AIS). This study was a randomized, controlled, single-blinded trial. Eighty AIS patients were split into two groups: the HUK and control groups. The HUK group was administered HUK and standard treatment, while the control group received standard treatment only. At admission and discharge, the National Institutes of Health Stroke Scale (NIHSS), Barthel Index (BI) and muscle strength were scored. The primary endpoint was the short-term outcomes of AIS patients under different treatments. The secondary endpoint was the degree of corticospinal tract fiber damage under different treatments. There was a significant improvement in the NIHSS Scale, BI and muscle strength scores in the HUK group compared with controls (Mann-Whitney U test; P < 0.05). Diffusion tensor tractography classification and intracranial arterial stenosis were independent predictors of short-term recovery by linear regression analysis. The changes in fractional anisotropy (FA) and apparent diffusion coefficient (ADC) decline rate were significantly smaller in the HUK group than in the control group ( P < 0.05). Vascular endothelial growth factor (VEGF) increased significantly after HUK treatment ( P < 0.05), and the VEGF change was negatively correlated with changes in ADC. HUK is beneficial for the outcome in AIS patients especially in motor function recovery. It may have protective effects on the corticospinal tract which is reflected by the reduction in the FA and ADC decline rates and increased VEGF expression. The study was registered on ClinicalTrials.gov (unique identifier: NCT04102956).
Collapse
Affiliation(s)
- Peifang Li
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
- Department of Neurology, Handan Central Hospital, Handan
| | - Honglin Lu
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
| | - Xiaoman Shi
- Department of Pediatrics, Affiliated Hospital of Hebei University, Baoding
| | - Jiajia Yan
- Department of Neurology, Cangzhou Integrated Traditional Chinese and Western Medicine Hospital, Cangzhou
| | - Lixia Zhou
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
| | - Jipeng Yang
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
| | - Binbin Wang
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
| | - Yanying Zhao
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
| | - Luji Liu
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
| | - Yipu Zhu
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
| | - Lei Xu
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
| | - Xiaoli Yang
- Department of Neurology, Hebei University of Engineering School of Medicine, Handan
| | - Xudong Su
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
| | - Yi Yang
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
| | - Tong Zhang
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
| | - Li Guo
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
| | - Xiaoyun Liu
- Department of Neurology, the Second Hospital of Hebei Medical University, Shijiazhuang
- Department of Neurology, the First Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
4
|
Popescu RG, Dinischiotu A, Soare T, Vlase E, Marinescu GC. Nicotinamide Mononucleotide (NMN) Works in Type 2 Diabetes through Unexpected Effects in Adipose Tissue, Not by Mitochondrial Biogenesis. Int J Mol Sci 2024; 25:2594. [PMID: 38473844 DOI: 10.3390/ijms25052594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/01/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024] Open
Abstract
Nicotinamide mononucleotide (NMN) has emerged as a promising therapeutic intervention for age-related disorders, including type 2 diabetes. In this study, we confirmed the previously observed effects of NMN treatment on glucose uptake and investigated its underlying mechanisms in various tissues and cell lines. Through the most comprehensive proteomic analysis to date, we discovered a series of novel organ-specific effects responsible for glucose uptake as measured by the IPGTT: adipose tissue growing (suggested by increased protein synthesis and degradation and mTOR proliferation signaling upregulation). Notably, we observed the upregulation of thermogenic UCP1, promoting enhanced glucose conversion to heat in intermuscular adipose tissue while showing a surprising repressive effect on mitochondrial biogenesis in muscle and the brain. Additionally, liver and muscle cells displayed a unique response, characterized by spliceosome downregulation and concurrent upregulation of chaperones, proteasomes, and ribosomes, leading to mildly impaired and energy-inefficient protein synthesis machinery. Furthermore, our findings revealed remarkable metabolic rewiring in the brain. This involved increased production of ketone bodies, downregulation of mitochondrial OXPHOS and TCA cycle components, as well as the induction of well-known fasting-associated effects. Collectively, our data elucidate the multifaceted nature of NMN action, highlighting its organ-specific effects and their role in improving glucose uptake. These findings deepen our understanding of NMN's therapeutic potential and pave the way for novel strategies in managing metabolic disorders.
Collapse
Affiliation(s)
- Roua Gabriela Popescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
- Independent Research Association, 012416 Bucharest, Romania
- Blue Screen SRL, 012416 Bucharest, Romania
| | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
| | - Teodoru Soare
- Pathology Department, Faculty of Veterinary Medicine, University of Agronomic Sciences and Veterinary Medicine of Bucharest, 050097 Bucharest, Romania
| | - Ene Vlase
- Animals Facility Laboratory, Cantacuzino National Institute for Medico-Military Research and Development, 013821 Bucharest, Romania
| | - George Cătălin Marinescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
- Independent Research Association, 012416 Bucharest, Romania
- Blue Screen SRL, 012416 Bucharest, Romania
| |
Collapse
|
5
|
Brusco I, Fialho MFP, Becker G, Brum ES, Favarin A, Marquezin LP, Serafini PT, Oliveira SM. Kinins and their B 1 and B 2 receptors as potential therapeutic targets for pain relief. Life Sci 2023; 314:121302. [PMID: 36535404 DOI: 10.1016/j.lfs.2022.121302] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Kinins are endogenous peptides that belong to the kallikrein-kinin system, which has been extensively studied for over a century. Their essential role in multiple physiological and pathological processes is demonstrated by activating two transmembrane G-protein-coupled receptors, the kinin B1 and B2 receptors. The attention is mainly given to the pathological role of kinins in pain transduction mechanisms. In the past years, a wide range of preclinical studies has amounted to the literature reinforcing the need for an updated review about the participation of kinins and their receptors in pain disorders. Here, we performed an extensive literature search since 2004, describing the historical progress and the current understanding of the kinin receptors' participation and its potential therapeutic in several acute and chronic painful conditions. These include inflammatory (mainly arthritis), neuropathic (caused by different aetiologies, such as cancer, multiple sclerosis, antineoplastic toxicity and diabetes) and nociplastic (mainly fibromyalgia) pain. Moreover, we highlighted the pharmacological actions and possible clinical applications of the kinin B1 and B2 receptor antagonists, kallikrein inhibitors or kallikrein-kinin system signalling pathways-target molecules in these different painful conditions. Notably, recent findings sought to elucidate mechanisms for guiding new and better drug design targeting kinin B1 and B2 receptors to treat a disease diversity. Since the kinin B2 receptor antagonist, Icatibant, is clinically used and well-tolerated by patients with hereditary angioedema gives us hope kinin receptors antagonists could be more robustly tested for a possible clinical application in the treatment of pathological pains, which present limited pharmacology management.
Collapse
Affiliation(s)
- Indiara Brusco
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Maria Fernanda Pessano Fialho
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Gabriela Becker
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Evelyne Silva Brum
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Amanda Favarin
- Laboratory of Neurotoxicity and Psychopharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Lara Panazzolo Marquezin
- Laboratory of Neurotoxicity and Psychopharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Patrick Tuzi Serafini
- Laboratory of Neurotoxicity and Psychopharmacology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Sara Marchesan Oliveira
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
6
|
Rodríguez-Massó SR, Erickson MA, Banks WA, Ulrich H, Martins AH. The Bradykinin B2 Receptor Agonist (NG291) Causes Rapid Onset of Transient Blood-Brain Barrier Disruption Without Evidence of Early Brain Injury. Front Neurosci 2021; 15:791709. [PMID: 34975388 PMCID: PMC8715084 DOI: 10.3389/fnins.2021.791709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Background: The blood-brain barrier (BBB) describes the brain's highly specialized capillaries, which form a dynamic interface that maintains central nervous system (CNS) homeostasis. The BBB supports the CNS, in part, by preventing the entry of potentially harmful circulating molecules into the brain. However, this specialized function is challenging for the development of CNS therapeutics. Several strategies to facilitate drug delivery into the brain parenchyma via disruption of the BBB have been proposed. Bradykinin has proven effective in disrupting mechanisms across the blood-tumor barrier. Unfortunately, bradykinin has limited therapeutic value because of its short half-life and the undesirable biological activity elicited by its active metabolites. Objective: To evaluate NG291, a stable bradykinin analog, with selective agonist activity on the bradykinin-B2 receptor and its ability to disrupt the BBB transiently. Methods: Sprague Dawley rats and CD-1 mice were subjected to NG291 treatment (either 50 or 100 μg/kg, intravenously). Time and dose-dependent BBB disruption were evaluated by histological analysis of Evans blue (EB) extravasation. Transcellular and paracellular BBB leakage were assessed by infiltration of 99mTc-albumin (66.5 KDa) and 14C-sucrose (340 Da) radiolabeled probes into the brains of CD-1 mice treated with NG291. NG291 influence on P-glycoprotein (P-gp) efflux pump activity was evaluated by quantifying the brain accumulation of 3H-verapamil, a known P-gp substrate, in CD-1 mice. Results: NG291-mediated BBB disruption was localized, dose-dependent, and reversible as measured by EB extravasation. 99mTc-albumin leakage was significantly increased by 50 μg/kg of NG291, whereas 100 μg/kg of NG291 significantly augmented both 14C-sucrose and 99mTc-albumin leakage. NG291 enhanced P-gp efflux transporter activity and was unable to increase brain uptake of the P-gp substrate pralidoxime. NG291 did not evoke significant short-term neurotoxicity, as it did not increase brain water content, the number of Fluoro-Jade C positive cells, or astrocyte activation. Conclusion: Our findings strongly suggest that NG291 increases BBB permeability by two different mechanisms in a dose-dependent manner and increases P-gp efflux transport. This increased permeability may facilitate the penetration into the brain of therapeutic candidates that are not P-gp substrates.
Collapse
Affiliation(s)
- Sergio R. Rodríguez-Massó
- Department of Pharmacology and Toxicology, University of Puerto Rico Medical Sciences Campus, San Juan, PR, United States
| | - Michelle A. Erickson
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Gerontology and Geriatric Medicine, Department of Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - William A. Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
- Division of Gerontology and Geriatric Medicine, Department of Medicine, School of Medicine, University of Washington, Seattle, WA, United States
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Antonio Henrique Martins
- Department of Pharmacology and Toxicology, University of Puerto Rico Medical Sciences Campus, San Juan, PR, United States
| |
Collapse
|
7
|
Alberto-Silva C, Portaro FCV, Kodama RT, Pantaleão HQ, Rangel M, Nihei KI, Konno K. Novel neuroprotective peptides in the venom of the solitary scoliid wasp Scolia decorata ventralis. J Venom Anim Toxins Incl Trop Dis 2021; 27:e20200171. [PMID: 34194483 PMCID: PMC8215932 DOI: 10.1590/1678-9199-jvatitd-2020-0171] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background Solitary wasp venoms may be a rich source of neuroactive substances, since their venoms are used for paralyzing preys. We have been exploring bioactive constituents of solitary wasp venoms and, in this study, the component profile of the venom from a solitary scoliid wasp, Scolia decorata ventralis, was investigated through a comprehensive analysis using LC-MS. Two peptides were synthesized, and their neuroprotective properties were evaluated. Methods A reverse-phase HPLC connected to ESI-MS was used for LC-MS analyses. Online mass fingerprinting was performed from TIC, and data-dependent tandem mass spectrometry gave the MS/MS spectra. The sequences of two major peptide components were determined by MALDI-TOF/TOF MS analysis, confirmed by solid phase synthesis. Using the synthetic peptides, biological activities were assessed. Cell integrity tests and neuroprotection analyzes using H2O2 as an oxidative stress inducer were performed for both peptides. Results Online mass fingerprinting revealed that the venom contains 123 components, and the MS/MS analysis resulted in 33 full sequences of peptide components. The two main peptides, α-scoliidine (DYVTVKGFSPLR) and β-scoliidine (DYVTVKGFSPLRKA), present homology with the bradykinin C-terminal. Despite this, both peptides did not behave as substrates or inhibitors of ACE, indicating that they do not interact with this metallopeptidase. In further studies, β-scoliidine, but not α -scoliidine, showed protective effects against oxidative stress-induced neurotoxicity in PC12 cells through integrity and metabolism cell assays. Interestingly, β-scoliidine has the extension of the KA dipeptide at the C-terminal in comparison with α-scoliidine. Conclusion Comprehensive LC-MS and MS/MS analyses from the Scolia decorata ventralis venom displayed the component profile of this venom. β-scoliidine showed an effective cytoprotective effect, probably due to the observed increase in the number of cells. This is the first report of solitary wasp venom peptides showing neuroprotective activity.
Collapse
Affiliation(s)
- Carlos Alberto-Silva
- Natural and Humanities Sciences Center, Experimental Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do Campo, SP, Brazil
| | | | | | - Halyne Queiroz Pantaleão
- Natural and Humanities Sciences Center, Experimental Morphophysiology Laboratory, Federal University of ABC (UFABC), São Bernardo do Campo, SP, Brazil
| | - Marisa Rangel
- Immunochemistry Laboratory, Butantan Institute, São Paulo, SP, Brazil
| | - Ken-Ichi Nihei
- Faculty of Agriculture, Utsunomiya University, Utsunomiya, Tochigi, Japan
| | - Katsuhiro Konno
- Institute of Natural Medicine, University of Toyama, Toyama, Toyama, Japan
| |
Collapse
|
8
|
Brusco I, Justino AB, Silva CR, Scussel R, Machado-de-Ávila RA, Oliveira SM. Inhibitors of angiotensin I converting enzyme potentiate fibromyalgia-like pain symptoms via kinin receptors in mice. Eur J Pharmacol 2021; 895:173870. [PMID: 33476653 DOI: 10.1016/j.ejphar.2021.173870] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 12/18/2020] [Accepted: 01/11/2021] [Indexed: 01/06/2023]
Abstract
Fibromyalgia is a potentially disabling chronic disease, characterized by widespread pain and a range of comorbidities such as hypertension. Among the mechanisms involved in fibromyalgia-like pain symptoms are kinins and their B1 and B2 receptors. Moreover, angiotensin I converting enzyme (ACE) inhibitors, commonly used as antihypertensive drugs, can enhance pain by blocking the degradation of peptides such as substance P and bradykinin, besides enhancing kinin receptors signalling. We investigated the effect of ACE inhibitors on reserpine-induced fibromyalgia-like pain symptoms and the involvement of kinins in this effect in mice. Nociceptive parameters (mechanical and cold allodynia and overt nociception) were evaluated after ACE inhibitors administration in mice previously treated with reserpine. The role of kinin B1 and B2 receptors was investigated using pharmacological antagonism. Additionally, bradykinin levels, as well as the activity of ACE and kininase I, were measured in the sciatic nerve, spinal cord and cerebral cortex of the mice. The ACE inhibitors enalapril and captopril enhanced reserpine-induced mechanical allodynia, and this increase was prevented by kinin B1 and B2 receptor antagonists. Substance P and bradykinin caused overt nociception and increased mechanical allodynia in animals treated with reserpine. Reserpine plus ACE inhibitors increased bradykinin-related peptide levels and inhibited ACE activity in pain modulation structures. Since hypertension is a frequent comorbidity affecting fibromyalgia patients, hypertension treatment with ACE inhibitors in these patients should be reviewed once this could enhance fibromyalgia-like pain symptoms. Thus, the treatment of hypertensive patients with fibromyalgia could include other classes of antihypertensive drugs, different from ACE inhibitors.
Collapse
Affiliation(s)
- Indiara Brusco
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Allisson Benatti Justino
- Graduate Program in Genetics and Biochemistry, Biotechnology Institute, Federal University of Uberlandia, Uberlandia, MG, Brazil
| | - Cássia Regina Silva
- Graduate Program in Genetics and Biochemistry, Biotechnology Institute, Federal University of Uberlandia, Uberlandia, MG, Brazil
| | - Rahisa Scussel
- Laboratory of Cellular and Molecular Biology, Health Sciences Academic Unit, University of Extreme South Catarinense, Criciuma, SC, Brazil
| | | | - Sara Marchesan Oliveira
- Graduate Program in Biological Sciences: Biochemistry Toxicology, Federal University of Santa Maria, Santa Maria, RS, Brazil.
| |
Collapse
|
9
|
Pillat MM, Oliveira-Giacomelli Á, das Neves Oliveira M, Andrejew R, Turrini N, Baranova J, Lah Turnšek T, Ulrich H. Mesenchymal stem cell-glioblastoma interactions mediated via kinin receptors unveiled by cytometry. Cytometry A 2021; 99:152-163. [PMID: 33438373 DOI: 10.1002/cyto.a.24299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/25/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022]
Abstract
Glioblastoma (GBM) is one of the most malignant and devastating brain tumors. The presence of highly therapy-resistant GBM cell subpopulations within the tumor mass, rapid invasion into brain tissues and reciprocal interactions with stromal cells in the tumor microenvironment contributes to an inevitable fatal prognosis for the patients. We highlight the most recent evidence of GBM cell crosstalk with mesenchymal stem cells (MSCs), which occurs either by direct cell-cell interactions via gap junctions and microtubules or cell fusion. MSCs and GBM paracrine interactions are commonly observed and involve cytokine signaling, regulating MSC tropism toward GBM, their intra-tumoral distribution, and immune system responses. MSC-promoted effects depending on their cytokine and receptor expression patterns are considered critical for GBM progression. MSC origin, tumor heterogeneity and plasticity may also determine the outcome of such interactions. Kinins and kinin-B1 and -B2 receptors play important roles in information flow between MSCs and GBM cells. Kinin-B1 receptor activity favors tumor migration and fusion of MSCs and GBM cells. Flow and image (tissue) cytometry are powerful tools to investigate GBM cell and MSC crosstalk and are applied to analyze and characterize several other cancer types.
Collapse
Affiliation(s)
- Micheli Mainardi Pillat
- Department of Microbiology and Parasitology, Health Sciences Center, Federal University of Santa Maria, Santa Maria, Brazil
| | | | | | - Roberta Andrejew
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Natalia Turrini
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Juliana Baranova
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Tamara Lah Turnšek
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Ljubljana, Slovenia
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Combination of Chemical and Neurotrophin Stimulation Modulates Neurotransmitter Receptor Expression and Activity in Transdifferentiating Human Adipose Stromal Cells. Stem Cell Rev Rep 2019; 15:851-863. [PMID: 31529274 DOI: 10.1007/s12015-019-09915-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adipose stromal cells are promising tools for clinical applications in regeneration therapies, due to their ease of isolation from tissue and its high yield; however, their ability to transdifferentiate into neural phenotypes is still a matter of controversy. Here, we show that combined chemical and neurotrophin stimulation resulted in neuron-like morphology and regulated expression and activity of several genes involved in neurogenesis and neurotransmission as well as ion currents mediated by NMDA and GABA receptors. Among them, expression patterns of genes coding for kinin-B1 and B2, α7 nicotinic, M1, M3 and M4 muscarinic acetylcholine, glutamatergic (AMPA2 and mGlu2), purinergic P2Y1 and P2Y4 and GABAergic (GABA-A, β3-subunit) receptors and neuronal nitric oxide synthase were up-regulated compared to levels of undifferentiated cells. Simultaneously, expression levels of P2X1, P2X4, P2X7 and P2Y6 purinergic and M5 muscarinic acetylcholine receptors were down-regulated. Agonist-induced activity levels of the studied receptor classes also augmented during neuronal transdifferentiation. Transdifferentiated cells expressed high levels of neuronal β3-tubulin, NF-H, NeuN and MAP-2 proteins as well as increased ASCL1, MYT1 and POU3F2 gene expression known to drive neuronal fate determination. The presented work contributes to a better understanding of transdifferentiation induced by neurotrophins for a prospective broad spectrum of medical applications.
Collapse
|
11
|
Oliveira MN, Breznik B, Pillat MM, Pereira RL, Ulrich H, Lah TT. Kinins in Glioblastoma Microenvironment. CANCER MICROENVIRONMENT 2019; 12:77-94. [PMID: 31420805 DOI: 10.1007/s12307-019-00229-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 07/29/2019] [Indexed: 12/16/2022]
Abstract
Tumour progression involves interactions among various cancer cell clones, including the cancer stem cell subpopulation and exogenous cellular components, termed cancer stromal cells. The latter include a plethora of tumour infiltrating immunocompetent cells, among which are also immuno-modulatory mesenchymal stem cells, which by vigorous migration to growing tumours and susequent transdifferentiation into various types of tumour-residing stromal cells, may either inhibit or support tumour progression. In the light of the scarce therapeutic options existing for the most malignant brain tumour glioblastoma, mesenchymal stem cells may represent a promising novel tool for cell therapy, e.g. drug delivery vectors. Here, we review the increasing number of reports on mutual interactions between mesenchymal stem cells and glioblastoma cells in their microenvironment. We particularly point out two novel aspects: the different responses of cancer cells to their microenvironmental cues, and to the signalling by kinin receptors that complement the immuno-modulating cytokine-signalling networks. Inflammatory glioblastoma microenvironment is characterised by increasing expression of kinin receptors during progressive glioma malignancy, thus making kinin signalling and kinins themselves rather important in this context. In general, their role in tumour microenvironment has not been explored so far. In addition, kinins also regulate blood brain barrier-related drug transfer as well as brain tumour angiogenesis. These studies support the on-going research on kinin antagonists as candidates in the development of anti-invasive agents for adjuvant glioblastoma therapy.
Collapse
Affiliation(s)
- Mona N Oliveira
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineus Prestes 748, São Paulo, SP, 05508-000, Brazil.,Jožef Stefan International Postgraduate School, Jamova, 39 1000, Ljubljana, Slovenia
| | - Barbara Breznik
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000, Ljubljana, Slovenia.
| | - Micheli M Pillat
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineus Prestes 748, São Paulo, SP, 05508-000, Brazil
| | - Ricardo L Pereira
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineus Prestes 748, São Paulo, SP, 05508-000, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, Av. Prof. Lineus Prestes 748, São Paulo, SP, 05508-000, Brazil
| | - Tamara T Lah
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000, Ljubljana, Slovenia.,Department of Biochemistry, Faculty of Chemistry and Chemical Engineering, University of Ljubljana, Večna pot 113, 1000, Ljubljana, Slovenia
| |
Collapse
|
12
|
Bradykinin B2 receptor is essential to running-induced cell proliferation in the adult mouse hippocampus. Brain Struct Funct 2018; 223:3901-3907. [DOI: 10.1007/s00429-018-1711-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 07/04/2018] [Indexed: 12/21/2022]
|
13
|
Gauberti M, Potzeha F, Vivien D, Martinez de Lizarrondo S. Impact of Bradykinin Generation During Thrombolysis in Ischemic Stroke. Front Med (Lausanne) 2018; 5:195. [PMID: 30018956 PMCID: PMC6037726 DOI: 10.3389/fmed.2018.00195] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/14/2018] [Indexed: 12/22/2022] Open
Abstract
Ischemic stroke is one of the leading causes of death and disability worldwide. Current medical management in the acute phase is based on the activation of the fibrinolytic cascade by intravenous injection of a plasminogen activator (such as tissue-type plasminogen activator, tPA) that promotes restauration of the cerebral blood flow and improves stroke outcome. Unfortunately, the use of tPA is associated with deleterious effects such as hemorrhagic transformation, symptomatic brain edema, and angioedema, which limit the efficacy of this therapeutic strategy. Preclinical and clinical evidence suggests that intravenous thrombolysis generates large amounts of bradykinin, a peptide with potent pro-inflammatory, and pro-edematous effects. This tPA-triggered generation of bradykinin could participate in the deleterious effects of thrombolysis and is a potential target to improve neurological outcome in tPA-treated patients. The present review aims at summarizing current evidence linking thrombolysis, bradykinin generation, and neurovascular damage.
Collapse
Affiliation(s)
- Maxime Gauberti
- Normandie Univ, UNICAEN, Institut National de la Santé et de la Recherche Médicale UMR-S U1237, "Physiopathology and Imaging of Neurological Disorders" PhIND, Caen, France.,Department of Diagnostic Imaging and Interventional Radiology, Centre Hospitalier Universitaire Caen Côte de Nacre, Caen, France
| | - Fanny Potzeha
- Normandie Univ, UNICAEN, Institut National de la Santé et de la Recherche Médicale UMR-S U1237, "Physiopathology and Imaging of Neurological Disorders" PhIND, Caen, France
| | - Denis Vivien
- Normandie Univ, UNICAEN, Institut National de la Santé et de la Recherche Médicale UMR-S U1237, "Physiopathology and Imaging of Neurological Disorders" PhIND, Caen, France.,Department of Clinical Research, Centre Hospitalier Universitaire Caen, Caen, France
| | - Sara Martinez de Lizarrondo
- Normandie Univ, UNICAEN, Institut National de la Santé et de la Recherche Médicale UMR-S U1237, "Physiopathology and Imaging of Neurological Disorders" PhIND, Caen, France
| |
Collapse
|
14
|
Activation of bradykinin B2 receptor induced the inflammatory responses of cytosolic phospholipase A 2 after the early traumatic brain injury. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2957-2971. [PMID: 29894755 DOI: 10.1016/j.bbadis.2018.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/02/2018] [Accepted: 06/07/2018] [Indexed: 12/17/2022]
Abstract
Phospholipase A2 is a known aggravator of inflammation and deteriorates neurological outcomes after traumatic brain injury (TBI), however the exact inflammatory mechanisms remain unknown. This study investigated the role of bradykinin and its receptor, which are known initial mediators within inflammation activation, as well as the mechanisms of the cytosolic phospholipase A2 (cPLA2)-related inflammatory responses after TBI. We found that cPLA2 and bradykinin B2 receptor were upregulated after a TBI. Rats treated with the bradykinin B2 receptor inhibitor LF 16-0687 exhibited significantly less cPLA2 expression and related inflammatory responses in the brain cortex after sustaining a controlled cortical impact (CCI) injury. Both the cPLA2 inhibitor and the LF16-0687 improved CCI rat outcomes by decreasing neuron death and reducing brain edema. The following TBI model utilized both primary astrocytes and primary neurons in order to gain further understanding of the inflammation mechanisms of the B2 bradykinin receptor and the cPLA2 in the central nervous system. There was a stronger reaction from the astrocytes as well as a protective effect of LF16-0687 after the stretch injury and bradykinin treatment. The protein kinase C pathway was thought to be involved in the B2 bradykinin receptor as well as the cPLA2-related inflammatory responses. Rottlerin, a Protein Kinase C (PKC) δ inhibitor, decreased the activity of the cPLA2 activity post-injury, and LF16-0687 suppressed both the PKC pathway and the cPLA2 activity within the astrocytes. These results indicated that the bradykinin B2 receptor-mediated pathway is involved in the cPLA2-related inflammatory response from the PKC pathway.
Collapse
|
15
|
Tu M, Liu H, Zhang R, Chen H, Mao F, Cheng S, Lu W, Du M. Analysis and Evaluation of the Inhibitory Mechanism of a Novel Angiotensin-I-Converting Enzyme Inhibitory Peptide Derived from Casein Hydrolysate. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:4139-4144. [PMID: 29637780 DOI: 10.1021/acs.jafc.8b00732] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Casein hydrolysates exert various biological activities, and the responsible functional peptides are being identified from them continuously. In this study, the tryptic casein hydrolysate was fractionated by an ultrafiltration membrane (3 kDa), and the peptides were identified by capillary electrophoresis-quadrupole-time-of-flight-tandem mass spectrometry. Meanwhile, in silico methods were used to analyze the toxicity, solubility, stability, and affinity between the peptides and angiotensin-I-converting enzyme (ACE). Finally, a new angiotensin-I-converting enzyme inhibitory (ACEI) peptide, EKVNELSK, derived from αs1-casein (fragment 35-42) was screened. The half maximal inhibitory concentration value of the peptide is 5.998 mM, which was determined by a high-performance liquid chromatography method. The Lineweaver-Burk plot indicated that this peptide is a mixed-type inhibitor against ACE. Moreover, Discovery Studio 2017 R2 software was adopted to perform molecular docking to propose the potential mechanisms underlying the ACEI activity of the peptide. These results indicated that EKVNELSK is a new ACEI peptide identified from casein hydrolysate.
Collapse
Affiliation(s)
- Maolin Tu
- School of Food Science and Technology, National Engineering Research Center of Seafood , Dalian Polytechnic University , Dalian , Liaoning 116034 , People's Republic of China
- Department of Food Science and Engineering, School of Chemistry and Chemical Engineering , Harbin Institute of Technology , Harbin , Heilongjiang 150001 , People's Republic of China
| | - Hanxiong Liu
- School of Food Science and Technology, National Engineering Research Center of Seafood , Dalian Polytechnic University , Dalian , Liaoning 116034 , People's Republic of China
| | - Ruyi Zhang
- School of Food Science and Technology, National Engineering Research Center of Seafood , Dalian Polytechnic University , Dalian , Liaoning 116034 , People's Republic of China
| | - Hui Chen
- School of Food Science and Technology, National Engineering Research Center of Seafood , Dalian Polytechnic University , Dalian , Liaoning 116034 , People's Republic of China
| | - Fengjiao Mao
- School of Food Science and Technology, National Engineering Research Center of Seafood , Dalian Polytechnic University , Dalian , Liaoning 116034 , People's Republic of China
| | - Shuzhen Cheng
- School of Food Science and Technology, National Engineering Research Center of Seafood , Dalian Polytechnic University , Dalian , Liaoning 116034 , People's Republic of China
- College of Food Science and Nutritional Engineering , China Agricultural University , Beijing 100083 , People's Republic of China
| | - Weihong Lu
- Department of Food Science and Engineering, School of Chemistry and Chemical Engineering , Harbin Institute of Technology , Harbin , Heilongjiang 150001 , People's Republic of China
| | - Ming Du
- School of Food Science and Technology, National Engineering Research Center of Seafood , Dalian Polytechnic University , Dalian , Liaoning 116034 , People's Republic of China
- Department of Food Science and Engineering, School of Chemistry and Chemical Engineering , Harbin Institute of Technology , Harbin , Heilongjiang 150001 , People's Republic of China
| |
Collapse
|
16
|
Soares DDM, Santos DR, Rummel C, Ott D, Melo MCC, Roth J, Calixto JB, Souza GEP. The relevance of kalikrein-kinin system via activation of B 2 receptor in LPS-induced fever in rats. Neuropharmacology 2017; 126:84-96. [PMID: 28826826 DOI: 10.1016/j.neuropharm.2017.08.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 08/05/2017] [Accepted: 08/11/2017] [Indexed: 12/21/2022]
Abstract
PURPOSE This study evaluated the involvement of endogenous kallikrein-kinin system and the bradykinin (BK) B1 and B2 receptors on LPS- induced fever and the POA cells involved in this response. MATERIAL AND METHODS Male Wistar rats received either i.v. (1 mg/kg), i.c.v. (20 nmol) or i.h. (2 nmol) injections of icatibant (B2 receptor antagonist) 30 or 60 min, respectively, before the stimuli. DALBK (B1 receptor antagonist) was given either 15min before BK (i.c.v.) or 30 min before LPS (i.v.). Captopril (5 mg/kg, sc.,) was given 1 h prior LPS or BK. Concentrations of BK and total kininogenon CSF, plasma and tissue kallikrein were evaluated. Rectal temperatures (rT) were assessed by telethermometry. Ca++ signaling in POA cells was performed in rat pup brain tissue microcultures. RESULTS Icatibant reduced LPS fever while, captopril exacerbated that response, an effect abolished by icatibant. Icatibant (i.h.) reduced fever to BK (i.h.) but not that induced by LPS (i.v.). BK increased intracellular calcium concentration in neurons and astrocytes. LPS increased levels of bradykinin, tissue kallikrein and total kininogen. BK (i.c.v.) increased rT and decreased tail skin temperature. Captopril potentiated BK-induced fever an effect abolished by icatibant. DALBK reduced the fever induced by BK. BK (i.c.v.) increased the CSF PGE2concentration. Effect abolished by indomethacin (i.p.). CONCLUSIONS LPS activates endogenous kalikrein-kinin system leading to production of BK, which by acting on B2-receptors of POA cells causes prostaglandin synthesis that in turn produces fever. Thus, a kinin B2-receptor antagonist that enters into the brain could constitute a new and interesting strategy to treat fever.
Collapse
Affiliation(s)
- Denis de Melo Soares
- Department of Medicament, Faculty of Pharmacy of Federal University of Bahia, Laboratory of Pharmacology, Ribeirão Preto, SP, Brazil.
| | - Danielle R Santos
- Pharmacology, Department of Physic and Chemistry, Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Christoph Rummel
- Veterinary Physiology, Faculty of Veterinary Medicine, Justus-Liebig-University of Giessen, Germany
| | - Daniela Ott
- Veterinary Physiology, Faculty of Veterinary Medicine, Justus-Liebig-University of Giessen, Germany
| | - Míriam C C Melo
- Pharmacology, Department of Physic and Chemistry, Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Joachim Roth
- Veterinary Physiology, Faculty of Veterinary Medicine, Justus-Liebig-University of Giessen, Germany
| | - João B Calixto
- Center of Innovation and Preclinical Research, Florianópolis, SC, Brazil
| | - Glória E P Souza
- Pharmacology, Department of Physic and Chemistry, Faculty of Pharmaceutical Sciences, University of São Paulo, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
17
|
Sestile CC, Maraschin JC, Gama VS, Zangrossi H, Graeff FG, Audi EA. Panicolytic-like action of bradykinin in the dorsal periaqueductal gray through μ-opioid and B2-kinin receptors. Neuropharmacology 2017; 123:80-87. [PMID: 28554847 DOI: 10.1016/j.neuropharm.2017.05.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/09/2017] [Accepted: 05/23/2017] [Indexed: 01/06/2023]
Abstract
A wealth of evidence has shown that opioid and kinin systems may control proximal defense in the dorsal periaqueductal gray matter (dPAG), a critical panic-associated area. Studies with drugs that interfere with serotonin-mediated neurotransmission suggest that the μ-opioid receptor (MOR) synergistically interacts with the 5-HT1A receptor in the dPAG to inhibit escape, a panic-related behavior. A similar inhibitory effect has also been reported after local administration of bradykinin (BK), which is blocked by the non-selective opioid receptor antagonist naloxone. The latter evidence, points to an interaction between BK and opioids in the dPAG. We further explored the existence of this interaction through the dPAG electrical stimulation model of panic. We also investigated whether intra-dPAG injection of captopril, an inhibitor of the angiotensin-converting enzyme (ACE) that also degrades BK, causes a panicolytic-like effect. Our results showed that intra-dPAG injection of BK inhibited escape performance in a dose-dependent way, and this panicolytic-like effect was blocked by the BK type 2 receptor (B2R) antagonist HOE-140, and by the selective MOR antagonist CTOP. Conversely, the panicolytic-like effect caused by local administration of the selective MOR agonist DAMGO was antagonized by pre-treatment with either CTOP or HOE-140, indicating cross-antagonism between MOR and B2R. Finally, intra-dPAG injection of captopril also impaired escape in a dose-dependent way, and this panicolytic-like effect was blocked by pretreatment with HOE-140, suggesting mediation by endogenous BK. The panicolytic-like effect of captopril indicates that the use of ACE inhibitors in the clinical management of panic disorder may be worth exploring.
Collapse
Affiliation(s)
- Caio César Sestile
- Department of Pharmacology and Therapeutics, State University of Maringá (UEM), Maringá, PR, Brazil.
| | | | - Vanessa Scalco Gama
- Department of Pharmacology and Therapeutics, State University of Maringá (UEM), Maringá, PR, Brazil
| | - Hélio Zangrossi
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP, Brazil; Institute of Neurosciences and Behavior (INeC), Ribeirão Preto, Brazil
| | | | - Elisabeth Aparecida Audi
- Department of Pharmacology and Therapeutics, State University of Maringá (UEM), Maringá, PR, Brazil.
| |
Collapse
|
18
|
Hopp S, Nolte MW, Stetter C, Kleinschnitz C, Sirén AL, Albert-Weissenberger C. Alleviation of secondary brain injury, posttraumatic inflammation, and brain edema formation by inhibition of factor XIIa. J Neuroinflammation 2017; 14:39. [PMID: 28219400 PMCID: PMC5319055 DOI: 10.1186/s12974-017-0815-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 02/08/2017] [Indexed: 11/15/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a devastating neurological condition and a frequent cause of permanent disability. Posttraumatic inflammation and brain edema formation, two pathological key events contributing to secondary brain injury, are mediated by the contact-kinin system. Activation of this pathway in the plasma is triggered by activated factor XII. Hence, we set out to study in detail the influence of activated factor XII on the abovementioned pathophysiological features of TBI. Methods Using a cortical cryogenic lesion model in mice, we investigated the impact of genetic deficiency of factor XII and inhibition of activated factor XII with a single bolus injection of recombinant human albumin-fused Infestin-4 on the release of bradykinin, the brain lesion size, and contact-kinin system-dependent pathological events. We determined protein levels of bradykinin, intracellular adhesion molecule-1, CC-chemokine ligand 2, and interleukin-1β by enzyme-linked immunosorbent assays and mRNA levels of genes related to inflammation by quantitative real-time PCR. Brain lesion size was determined by tetrazolium chloride staining. Furthermore, protein levels of the tight junction protein occludin, integrity of the blood-brain barrier, and brain water content were assessed by Western blot analysis, extravasated Evans Blue dye, and the wet weight-dry weight method, respectively. Infiltration of neutrophils and microglia/activated macrophages into the injured brain lesions was quantified by immunohistological stainings. Results We show that both genetic deficiency of factor XII and inhibition of activated factor XII in mice diminish brain injury-induced bradykinin release by the contact-kinin system and minimize brain lesion size, blood-brain barrier leakage, brain edema formation, and inflammation in our brain injury model. Conclusions Stimulation of bradykinin release by activated factor XII probably plays a prominent role in expanding secondary brain damage by promoting brain edema formation and inflammation. Pharmacological blocking of activated factor XII could be a useful therapeutic principle in the treatment of TBI-associated pathologic processes by alleviating posttraumatic inflammation and brain edema formation.
Collapse
Affiliation(s)
- Sarah Hopp
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany.,Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Strasse 11, Würzburg, Germany
| | | | - Christian Stetter
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Strasse 11, Würzburg, Germany
| | - Christoph Kleinschnitz
- Department of Neurology, University Hospital Würzburg, Würzburg, Germany.,Department of Neurology, University Duisburg-Essen, Essen, Germany
| | - Anna-Leena Sirén
- Department of Neurosurgery, University Hospital Würzburg, Josef-Schneider-Strasse 11, Würzburg, Germany
| | | |
Collapse
|
19
|
Potentiation of Paclitaxel-Induced Pain Syndrome in Mice by Angiotensin I Converting Enzyme Inhibition and Involvement of Kinins. Mol Neurobiol 2016; 54:7824-7837. [PMID: 27844290 DOI: 10.1007/s12035-016-0275-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/30/2016] [Indexed: 10/20/2022]
Abstract
Paclitaxel is a chemotherapeutic agent used to treat solid tumours. However, it causes an acute and neuropathic pain syndrome that limits its use. Among the mechanisms involved in neuropathic pain caused by paclitaxel is activation of kinin receptors. Angiotensin converting enzyme (ACE) inhibitors can enhance kinin receptor signalling. The goal of this study was to evaluate the role of kinins on paclitaxel-associated acute pain syndromes (P-APS) and the effect of ACE inhibition on P-APS and paclitaxel-associated chronic peripheral neuropathy (P-CPN) in mice. Herein, we show that paclitaxel caused mechanical allodynia and spontaneous nociceptive behaviour that was reduced by antagonists of kinin receptors B1 (DALBk and SSR240612) and B2 (Hoe140 and FR173657). Moreover, enalapril (an ACE inhibitor) enhanced the mechanical allodynia induced by a low dose of paclitaxel. Likewise, paclitaxel injection inhibited ACE activity and increased the expressions of B1 and B2 receptors and bradykinin-related peptides levels in peripheral tissue. Together, our data support the involvement of kinin receptors in the P-APS and suggest kinin receptor antagonists to treat this syndrome. Because hypertension is the most frequent comorbidity affecting cancer patients, treatment of hypertension with ACE inhibitors in patients undergoing paclitaxel chemotherapy should be reviewed, since this could enhance the P-APS and P-CPN.
Collapse
|
20
|
dos Anjos LC, Gomes FMM, do Couto LL, Mourão CA, Moreira KG, Silva LP, Mortari MR. Anxiolytic activity and evaluation of potentially adverse effects of a bradykinin-related peptide isolated from a social wasp venom. Life Sci 2016; 149:153-9. [PMID: 26898126 DOI: 10.1016/j.lfs.2016.02.063] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 02/12/2016] [Accepted: 02/16/2016] [Indexed: 01/04/2023]
Abstract
Anxiety disorders are major health problems in terms of costs stemming from sick leave, disabilities, healthcare and premature mortality. Despite the availability of classic anxiolytics, some anxiety disorders are still resistant to treatment, with higher rates of adverse effects. In this respect, several toxins isolated from arthropod venoms are useful in identifying new compounds to treat neurological disorders, particularly pathological anxiety. Thus, the aims of this study were to identify and characterize an anxiolytic peptide isolated from the venom of the social wasp Polybia paulista. The peptide was identified as Polisteskinin R, with nominal molecular mass [M+H](+)=1301Da and primary structure consisting of Ala-Arg-Arg-Pro-Pro-Gly-Phe-Thr-Pro-Phe-Arg-OH. The anxiolytic effect was tested using the elevated plus maze test. Moreover, adverse effects on the spontaneous behavior and motor coordination of animals were assessed using the open field and rotarod tests. Polisteskinin R induced a dose-dependent anxiolytic effect. Animals treated with the peptide and diazepam spent significantly more time into the open arms when compared to the groups treated with the vehicle and pentylenetetrazole. No significant differences in spontaneous behavior or motor coordination were observed between the groups, showing that the peptide was well tolerated. The interaction by agonists in both known BK receptors induces a variability of physiological effects; Polisteskinin R can act on these receptors, inducing modulatory activity and thus, attenuating anxiety behaviors. The results of this study demonstrated that the compound Polisteskinin R exerted potent anxiolytic effects and its analogues are promising candidates for experimental pharmacology.
Collapse
Affiliation(s)
- Lilian Carneiro dos Anjos
- Neuropharmacology Laboratory, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, Brazil; Graduate Program of Animal Biology, University of Brasília, Brasília, DF, Brazil
| | - Flávia Maria Medeiros Gomes
- Neuropharmacology Laboratory, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, Brazil; Graduate Program of Animal Biology, University of Brasília, Brasília, DF, Brazil
| | - Lucianna Lopes do Couto
- Neuropharmacology Laboratory, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, Brazil
| | - Cecília Alves Mourão
- Neuropharmacology Laboratory, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, Brazil
| | | | - Luciano Paulino Silva
- Graduate Program of Animal Biology, University of Brasília, Brasília, DF, Brazil; Laboratory of Mass Spectrometry and Laboratory of Nanobiotechnology, Embrapa Genetic Resources and Biotechnology, Brasília, DF, Brazil
| | - Márcia Renata Mortari
- Neuropharmacology Laboratory, Department of Physiological Sciences, Institute of Biological Sciences, University of Brasília, Brasília, DF, Brazil; Graduate Program of Animal Biology, University of Brasília, Brasília, DF, Brazil.
| |
Collapse
|
21
|
Nascimento IC, Glaser T, Nery AA, Pillat MM, Pesquero JB, Ulrich H. Kinin-B1 and B2 receptor activity in proliferation and neural phenotype determination of mouse embryonic stem cells. Cytometry A 2015; 87:989-1000. [PMID: 26243460 DOI: 10.1002/cyto.a.22726] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The kinins bradykinin and des-arg(9) -bradykinin cleaved from kininogen precursors by kallikreins exert their biological actions by stimulating kinin-B2 and B1 receptors, respectively. In vitro models of neural differentiation such as P19 embryonal carcinoma cells and neural progenitor cells have suggested the involvement of B2 receptors in neural differentiation and phenotype determination; however, the involvement of B1 receptors in these processes has not been established. Here, we show that B1 and B2 receptors are differentially expressed in mouse embryonic E14Tg2A stem cells undergoing neural differentiation. Proliferation and differentiation assays, performed in the presence of receptor subtype-selective agonists and antagonists, revealed that B1 receptor activity is required for the proliferation of embryonic and differentiating cells as well as for neuronal maturation at later stages of differentiation, while the B2 receptor acts on neural phenotype choice, promoting neurogenesis over gliogenesis. Besides the elucidation of bradykinin functions in an in vitro model reflecting early embryogenesis and neurogenesis, this study contributes to the understanding of B1 receptor functions in this process.
Collapse
Affiliation(s)
- Isis C Nascimento
- Departamento de Neurologia e Neurocirurgia, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Talita Glaser
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Arthur A Nery
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - Micheli M Pillat
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| | - João B Pesquero
- Departamento de Biofísica, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
22
|
Assis DM, Juliano L, Paschoalin T, Kouyoumdjian M, Calixto JB, Santos RAS, Pertinhez TA, Gauthier F, Moreau T, Blaber M, Juliano MA. Pharmacological Activities and Hydrolysis by Peptidases of [Phospho-Ser(6)]-Bradykinin (pS(6)-BK). Biochem Pharmacol 2015; 97:203-14. [PMID: 26235942 DOI: 10.1016/j.bcp.2015.07.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 07/27/2015] [Indexed: 12/27/2022]
Abstract
Phosphorylated kininogen and some of its fragments containing serine phosphorylated bradykinin ([pS(6)]-Bk) were identified in human serum and plasma by a phosphoproteomic approach. We report the kininogenase ability of human tissue and plasma kallikreins and tryptase to generate [pS(6)]-Bk or Lys-[pS(6)]-Bk having as substrate the synthetic human kininogen fluorescent fragment Abz-MISLMKRPPGF[pS(386)]PFRSSRI-NH2. The pharmacological assays of [pS(6)]-Bk showed it as a full B2 bradykinin receptor agonist in smooth muscle, it produces a portal liver hypertensive response in rat and mouse paw edema that lasts longer than Bk. The rat hypotensive response to infusions of Bk is greater than that of [pS(6)]Bk, both if injected through femoral vein or aorta. [pS(6)]-Bk was more resistant than Bk to kininase digestion performed with angiotensin converting enzyme, neprilysin, thimet oligopeptidase, aminopeptidase P and carboxypeptidase M. (1)H-NMR experiments indicated that [pS(6)]-Bk has lower flexibility, with the pS(6)-P(7) bond restricted to the trans conformation, and can explain [pS(6)]-Bk resistance to hydrolysis. In conclusion, [pS(6)]-Bk presenting lower activity than Bk, with longer lasting effects and being slowly released by kininogenases from synthetic Abz-MISLMKRPPGF[pS(386)]PFRSSRI-NH2, suggests that phosphorylation of the kininogens can be an efficient kallikrein-kinin system regulator.
Collapse
Affiliation(s)
- Diego M Assis
- Department of Biophysics, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo/SP, Brazil
| | - Luiz Juliano
- Department of Biophysics, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo/SP, Brazil.
| | - Thaysa Paschoalin
- Department of Biophysics, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo/SP, Brazil
| | - Maria Kouyoumdjian
- Department of Biochemistry, Biophysics and Medicine, São Paulo Federal University, São Paulo/SP, Brazil
| | - Joao B Calixto
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis/SC, Brazil
| | - Robson A S Santos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte/MG, Brazil
| | - Thelma A Pertinhez
- Transfusion Medicine Unit, Arcispedale Santa Maria Nuova-IRCCS, Reggio Emilia, Italy
| | | | | | - Michael Blaber
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Maria A Juliano
- Department of Biophysics, Escola Paulista de Medicina, Federal University of São Paulo, São Paulo/SP, Brazil.
| |
Collapse
|