1
|
Li J, Zeng S, Zhang E, Chen L, Jiang J, Li J. Spatial metabolomics to discover hypertrophic scar relevant metabolic alterations and potential therapeutic strategies: A preliminary study. Bioorg Chem 2024; 153:107873. [PMID: 39383811 DOI: 10.1016/j.bioorg.2024.107873] [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: 12/19/2023] [Revised: 08/22/2024] [Accepted: 10/04/2024] [Indexed: 10/11/2024]
Abstract
Spatially mapping the metabolic remodeling of hypertrophic scar and surrounding normal skin tissues has the potential to enhance our comprehension of scar formation and aid in the advancement of therapeutic interventions. In this study, we employed matrix-assisted laser desorption/ionization (MALDI), a mass spectrometry imaging technique, to visualize the hierarchical distribution of metabolites within sections of hypertrophic scar and surrounding normal skin tissues. A comprehensive analysis identified a total of 1631 metabolites in these tissues. The top four classes that were identified included benzene and substituted derivatives, heterocyclic compounds, amino acids and its metabolites, and glycerophospholipids. In hypertrophic scar tissues, 22 metabolites were upregulated and 66 metabolites were downregulated. MetaboAnalyst pathway analysis indicated that glycerophospholipid metabolism was primarily associated with these altered 88 metabolites. Subsequently, six metabolites were selected, their spatial characteristics were analyzed, and they were individually added to the cell culture medium of primary hypertrophic scar fibroblasts. The preliminary findings of this study demonstrate that specific concentrations of 1-pyrrolidinecarboxamide, 2-benzylideneheptanal, glycerol trioleate, Lyso-PAF C-16, and moxonidine effectively inhibited the expressions of COL1A1, COL1A2, COL3A1, and ACTA2. These bioactive metabolites exhibit mild and non-toxic properties, along with favorable pharmacokinetics and pharmacodynamics, making them promising candidates for drug development. Consequently, this research offers novel therapeutic insights for hypertrophic scar treatment.
Collapse
Affiliation(s)
- Jingyun Li
- Nanjing Women and Children's Healthcare Institute, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Siqi Zeng
- Department of Plastic & Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Enyuan Zhang
- Department of Plastic & Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Ling Chen
- Department of Plastic & Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Jingbin Jiang
- Department of Plastic & Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China
| | - Jun Li
- Department of Plastic & Cosmetic Surgery, Women's Hospital of Nanjing Medical University (Nanjing Women and Children's Healthcare Hospital), 123rd Tianfei Street, Mochou Road, Nanjing 210004, China.
| |
Collapse
|
2
|
Wang B, Mou H, Liu M, Ran Z, Li X, Li J, Ou Y. Multiomics characteristics of neurogenesis-related gene are dysregulated in tumor immune microenvironment. NPJ Genom Med 2021; 6:37. [PMID: 34059678 PMCID: PMC8166819 DOI: 10.1038/s41525-021-00202-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 05/05/2021] [Indexed: 11/09/2022] Open
Abstract
The success of immunotherapy was overshadowed by its low response rate, and the hot or cold tumor microenvironment was reported to be responsible for it. However, due to the lack of an appropriate method, it is still a huge challenge for researchers to understand the molecular differences between hot and cold tumor microenvironments. Further research is needed to gain deeper insight into the molecular characteristics of the hot/cold tumor microenvironment. A large-scale clinical cohort and single-cell RNA-seq technology were used to identify the molecular characteristics of inflamed or noninflamed tumors. With single-cell RNA sequencing technology, we provided a novel method to dissect the tumor microenvironment into a hot/cold tumor microenvironment to help us understand the molecular differences between hot and cold tumor microenvironments. Compared with cold tumors, hot tumors highly expressed B cell-related genes, such as MS4A1 and CXCR5, neurogenesis-related miRNA such as MIR650, and immune molecule-related lncRNA such as MIR155HG and LINC00426. In cold tumors, the expression of genes related to multiple biological processes, such as the neural system, was significantly upregulated, and methylome analysis indicated that the promoter methylation level of genes related to neurogenesis was significantly reduced. Finally, we investigated the pan-cancer prognostic value of the cold/hot microenvironment and performed pharmacogenomic analysis to predict potential drugs that may have the potential to convert the cold microenvironment into a hot microenvironment. Our study reveals the multiomics characteristics of cold/hot microenvironments. These molecular characteristics may contribute to the understanding of immune exclusion and the development of microenvironment-targeted therapy.
Collapse
Affiliation(s)
- Ben Wang
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hai Mou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mengmeng Liu
- Anhui No. 2 Provincial People's Hospital, Hefei, China
| | - Zhujie Ran
- School of Public Health and Community Medicine, Wenzhou Medical University, Wenzhou, China
| | - Xin Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Li
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunsheng Ou
- Department of Orthopedics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
3
|
Bousquet P, Hudson A, García-Sevilla JA, Li JX. Imidazoline Receptor System: The Past, the Present, and the Future. Pharmacol Rev 2020; 72:50-79. [PMID: 31819014 DOI: 10.1124/pr.118.016311] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Imidazoline receptors historically referred to a family of nonadrenergic binding sites that recognize compounds with an imidazoline moiety, although this has proven to be an oversimplification. For example, none of the proposed endogenous ligands for imidazoline receptors contain an imidazoline moiety but they are diverse in their chemical structure. Three receptor subtypes (I1, I2, and I3) have been proposed and the understanding of each has seen differing progress over the decades. I1 receptors partially mediate the central hypotensive effects of clonidine-like drugs. Moxonidine and rilmenidine have better therapeutic profiles (fewer side effects) than clonidine as antihypertensive drugs, thought to be due to their higher I1/α 2-adrenoceptor selectivity. Newer I1 receptor agonists such as LNP599 [3-chloro-2-methyl-phenyl)-(4-methyl-4,5-dihydro-3H-pyrrol-2-yl)-amine hydrochloride] have little to no activity on α 2-adrenoceptors and demonstrate promising therapeutic potential for hypertension and metabolic syndrome. I2 receptors associate with several distinct proteins, but the identities of these proteins remain elusive. I2 receptor agonists have demonstrated various centrally mediated effects including antinociception and neuroprotection. A new I2 receptor agonist, CR4056 [2-phenyl-6-(1H-imidazol-1yl) quinazoline], demonstrated clear analgesic activity in a recently completed phase II clinical trial and holds great promise as a novel I2 receptor-based first-in-class nonopioid analgesic. The understanding of I3 receptors is relatively limited. Existing data suggest that I3 receptors may represent a binding site at the Kir6.2-subtype ATP-sensitive potassium channels in pancreatic β-cells and may be involved in insulin secretion. Despite the elusive nature of their molecular identities, recent progress on drug discovery targeting imidazoline receptors (I1 and I2) demonstrates the exciting potential of these compounds to elicit neuroprotection and to treat various disorders such as hypertension, metabolic syndrome, and chronic pain.
Collapse
Affiliation(s)
- Pascal Bousquet
- Faculty of Medicine, University of Strasbourg, Strasbourg, France (P.B.); Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada (A.H.); Laboratory of Neuropharmacology, University Research Institute on Health Sciences, University of the Balearic Islands, Palma de Malllorca, Spain (J.A.G.-S.); and Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York (J.-X.L.)
| | - Alan Hudson
- Faculty of Medicine, University of Strasbourg, Strasbourg, France (P.B.); Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada (A.H.); Laboratory of Neuropharmacology, University Research Institute on Health Sciences, University of the Balearic Islands, Palma de Malllorca, Spain (J.A.G.-S.); and Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York (J.-X.L.)
| | - Jesús A García-Sevilla
- Faculty of Medicine, University of Strasbourg, Strasbourg, France (P.B.); Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada (A.H.); Laboratory of Neuropharmacology, University Research Institute on Health Sciences, University of the Balearic Islands, Palma de Malllorca, Spain (J.A.G.-S.); and Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York (J.-X.L.)
| | - Jun-Xu Li
- Faculty of Medicine, University of Strasbourg, Strasbourg, France (P.B.); Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada (A.H.); Laboratory of Neuropharmacology, University Research Institute on Health Sciences, University of the Balearic Islands, Palma de Malllorca, Spain (J.A.G.-S.); and Department of Pharmacology and Toxicology, University at Buffalo, Buffalo, New York (J.-X.L.)
| |
Collapse
|
4
|
Djikic T, Vucicevic J, Laurila J, Radi M, Veljkovic N, Xhaard H, Nikolic K. Deciphering Imidazoline Off‐targets by Fishing in the Class A of GPCR field. Mol Inform 2020; 39:e1900165. [DOI: 10.1002/minf.201900165] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 02/20/2020] [Indexed: 12/13/2022]
Affiliation(s)
- Teodora Djikic
- Department of Pharmaceutical ChemistryFaculty of PharmacyUniversity of Belgrade Vojvode Stepe 450 11000 Belgrade Serbia
| | - Jelica Vucicevic
- Department of Pharmaceutical ChemistryFaculty of PharmacyUniversity of Belgrade Vojvode Stepe 450 11000 Belgrade Serbia
| | - Jonne Laurila
- Research Center for Integrative Physiology and Pharmacology, Institute of BiomedicineUniversity of Turku FI-20014 Turun yliopisto, Turku Finland
| | - Marco Radi
- Dipartimento di Scienze degli Alimenti e del FarmacoUniversità degli Studi di Parma Viale delle Scienze, 27/A 43124 Parma Italy
| | - Nevena Veljkovic
- Laboratory for bioinformatics and computational chemistry, Institute of Nuclear Sciences VincaUniversity of Belgrade Mihaila Petrovica Alasa 14 11001 Belgrade Serbia
| | - Henri Xhaard
- Division of Pharmaceutical Chemistry, Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of PharmacyUniversity of Helsinki P.O. Box 56 FI-00014 Helsinki Finland
| | - Katarina Nikolic
- Department of Pharmaceutical ChemistryFaculty of PharmacyUniversity of Belgrade Vojvode Stepe 450 11000 Belgrade Serbia
| |
Collapse
|
5
|
Cobos-Puc L, Aguayo-Morales H. Cardiovascular Effects Mediated by Imidazoline Drugs: An Update. Cardiovasc Hematol Disord Drug Targets 2019; 19:95-108. [PMID: 29962350 DOI: 10.2174/1871529x18666180629170336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/05/2017] [Accepted: 04/18/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE Clonidine is a centrally acting antihypertensive drug. Hypotensive effect of clonidine is mediated mainly by central α2-adrenoceptors and/or imidazoline receptors located in a complex network of the brainstem. Unfortunately, clonidine produces side effects such as sedation, mouth dry, and depression. Moxonidine and rilmenidine, compounds of the second generation of imidazoline drugs, with fewer side effects, display a higher affinity for the imidazoline receptors compared with α2-adrenoceptors. The antihypertensive action of these drugs is due to inhibition of the sympathetic outflow primarily through central I1-imidazoline receptors in the RVLM, although others anatomical sites and mechanisms/receptors are involved. Agmatine is regarded as the endogenous ligand for imidazoline receptors. This amine modulates the cardiovascular function. Indeed, when administered in the RVLM mimics the hypotension of clonidine. RESULTS Recent findings have shown that imidazoline drugs also exert biological response directly on the cardiovascular tissues, which can contribute to their antihypertensive response. Currently, new imidazoline receptors ligands are in development. CONCLUSION In the present review, we provide a brief update on the cardiovascular effects of clonidine, moxonidine, rilmenidine, and the novel imidazoline agents since representing an important therapeutic target for some cardiovascular diseases.
Collapse
Affiliation(s)
- Luis Cobos-Puc
- Department of Pharmacology, Faculty of Chemistry, Autonomous University of Coahuila, Saltillo, Mexico
| | - Hilda Aguayo-Morales
- Department of Pharmacology, Faculty of Chemistry, Autonomous University of Coahuila, Saltillo, Mexico
| |
Collapse
|
6
|
Obergasteiger J, Frapporti G, Pramstaller PP, Hicks AA, Volta M. A new hypothesis for Parkinson's disease pathogenesis: GTPase-p38 MAPK signaling and autophagy as convergence points of etiology and genomics. Mol Neurodegener 2018; 13:40. [PMID: 30071902 PMCID: PMC6090926 DOI: 10.1186/s13024-018-0273-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 07/19/2018] [Indexed: 02/07/2023] Open
Abstract
The combination of genetics and genomics in Parkinson´s disease has recently begun to unveil molecular mechanisms possibly underlying disease onset and progression. In particular, catabolic processes such as autophagy have been increasingly gaining relevance as post-mortem evidence and experimental models suggested a participation in neurodegeneration and alpha-synuclein Lewy body pathology. In addition, familial Parkinson´s disease linked to LRRK2 and alpha-synuclein provided stronger correlation between etiology and alterations in autophagy. More detailed cellular pathways are proposed and genetic risk factors that associate with idiopathic Parkinson´s disease provide further clues in dissecting contributions of single players. Nevertheless, the fine-tuning of these processes remains elusive, as the initial stages of the pathways are not yet clarified.In this review, we collect literature evidence pointing to autophagy as the common, downstream target of Parkinsonian dysfunctions and augment current knowledge on the factors that direct the subsequent steps. Cell and molecular biology evidence indicate that p38 signaling underlies neurodegeneration and autoptic observations suggest a participation in neuropathology. Moreover, alpha-synuclein and LRRK2 also appear involved in the p38 pathway with additional roles in the regulation of GTPase signaling. Small GTPases are critical modulators of p38 activation and thus, their functional interaction with aSyn and LRRK2 could explain much of the detailed mechanics of autophagy in Parkinson´s disease.We propose a novel hypothesis for a more comprehensive working model where autophagy is controlled by upstream pathways, such as GTPase-p38, that have been so far underexplored in this context. In addition, etiological factors (LRRK2, alpha-synuclein) and risk loci might also combine in this common mechanism, providing a powerful experimental setting to dissect the cause of both familial and idiopathic disease.
Collapse
Affiliation(s)
- Julia Obergasteiger
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Giulia Frapporti
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Peter P. Pramstaller
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
- Department of Neurology, General Central Hospital, Via Böhler 5, 39100 Bolzano, Italy
- Department of Neurology, University of Lübeck, Ratzeburger Allee, 23538 Lübeck, Germany
| | - Andrew A. Hicks
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| | - Mattia Volta
- Institute for Biomedicine, Eurac Research – Affiliated Institute of the University of Lübeck, Via Galvani 31, 39100 Bolzano, Italy
| |
Collapse
|
7
|
Vucicevic J, Srdic-Rajic T, Pieroni M, Laurila JMM, Perovic V, Tassini S, Azzali E, Costantino G, Glisic S, Agbaba D, Scheinin M, Nikolic K, Radi M, Veljkovic N. A combined ligand- and structure-based approach for the identification of rilmenidine-derived compounds which synergize the antitumor effects of doxorubicin. Bioorg Med Chem 2016; 24:3174-83. [PMID: 27265687 DOI: 10.1016/j.bmc.2016.05.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2016] [Revised: 05/17/2016] [Accepted: 05/20/2016] [Indexed: 10/21/2022]
Abstract
The clonidine-like central antihypertensive agent rilmenidine, which has high affinity for I1-type imidazoline receptors (I1-IR) was recently found to have cytotoxic effects on cultured cancer cell lines. However, due to its pharmacological effects resulting also from α2-adrenoceptor activation, rilmenidine cannot be considered a suitable anticancer drug candidate. Here, we report the identification of novel rilmenidine-derived compounds with anticancer potential and devoid of α2-adrenoceptor effects by means of ligand- and structure-based drug design approaches. Starting from a large virtual library, eleven compounds were selected, synthesized and submitted to biological evaluation. The most active compound 5 exhibited a cytotoxic profile similar to that of rilmenidine, but without appreciable affinity to α2-adrenoceptors. In addition, compound 5 significantly enhanced the apoptotic response to doxorubicin, and may thus represent an important tool for the development of better adjuvant chemotherapeutic strategies for doxorubicin-insensitive cancers.
Collapse
Affiliation(s)
- Jelica Vucicevic
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Tatjana Srdic-Rajic
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - Marco Pieroni
- P4T Group, Dipartimento di Farmacia, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| | - Jonne M M Laurila
- Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland
| | - Vladimir Perovic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences Vinca, University of Belgrade, POB 522, Mihaila Petrovica Alasa 14, 11001 Belgrade, Serbia
| | - Sabrina Tassini
- P4T Group, Dipartimento di Farmacia, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| | - Elisa Azzali
- P4T Group, Dipartimento di Farmacia, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| | - Gabriele Costantino
- P4T Group, Dipartimento di Farmacia, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy
| | - Sanja Glisic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences Vinca, University of Belgrade, POB 522, Mihaila Petrovica Alasa 14, 11001 Belgrade, Serbia
| | - Danica Agbaba
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia
| | - Mika Scheinin
- Department of Pharmacology, Drug Development and Therapeutics, University of Turku, Kiinamyllynkatu 10, FI-20520 Turku, Finland; Unit of Clinical Pharmacology, Turku University Hospital, Turku, Finland
| | - Katarina Nikolic
- Institute of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11000 Belgrade, Serbia.
| | - Marco Radi
- P4T Group, Dipartimento di Farmacia, Università degli Studi di Parma, Viale delle Scienze, 27/A, 43124 Parma, Italy.
| | - Nevena Veljkovic
- Center for Multidisciplinary Research, Institute of Nuclear Sciences Vinca, University of Belgrade, POB 522, Mihaila Petrovica Alasa 14, 11001 Belgrade, Serbia.
| |
Collapse
|
8
|
Srdic-Rajic T, Nikolic K, Cavic M, Djokic I, Gemovic B, Perovic V, Veljkovic N. Rilmenidine suppresses proliferation and promotes apoptosis via the mitochondrial pathway in human leukemic K562 cells. Eur J Pharm Sci 2016; 81:172-80. [DOI: 10.1016/j.ejps.2015.10.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 10/12/2015] [Accepted: 10/22/2015] [Indexed: 12/20/2022]
|
9
|
Keller B, García-Sevilla JA. Immunodetection and subcellular distribution of imidazoline receptor proteins with three antibodies in mouse and human brains: Effects of treatments with I1- and I2-imidazoline drugs. J Psychopharmacol 2015; 29:996-1012. [PMID: 26038110 DOI: 10.1177/0269881115586936] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Various imidazoline receptor (IR) proteins have been proposed to mediate the effects of selective I1- and I2-IR drugs. However, the association of these IR-binding proteins with classic I1- and I2-radioligand binding sites remains somewhat controversial. In this study, three IR antibodies (anti-NISCH and anti-nischarin for I1-IRs; and anti-IRBP for I1/I2-IRs) were used to immunodetect, characterize and compare IR protein patterns in brain (mouse and human; total homogenate, subcellular fractionation, grey and white matter) and some cell systems (neurones, astrocytes, human platelets). Various immunoreactive IRs (specific molecular weight bands coincidently detected with the different antibodies) were related to I1-IR (167 kDa, 105/115 kDa and 85 kDa proteins) or I2-IR (66 kDa, 45 kDa and 30 kDa proteins) types. The biochemical characterization of cortical 167 kDa protein, localized in the membrane/cytosol but not in the nucleus, indicated that this I1-IR also forms part of higher order nischarin-related complexes. The contents of I1-IR (167 kDa, 105/115 kDa, and 85 kDa) proteins in mouse brain cortex were upregulated by treatment with I1-drugs (moxonidine, efaroxan) but not with I2-drugs (BU-224, LSL 61122). Conversely, the contents of I2-IR (66 kDa, 45 kDa and 30 kDa) proteins in mouse brain cortex were modulated by treatment with I2-drugs (decreases after BU-224 and LSL 61122, and increases after idazoxan) but not with I1-drugs (with the exception of moxonidine). These findings further indicate that brain immunoreactive IR proteins exist in multiple forms that can be grouped in the already known I1- and I2-IR types, which are expressed both in neurones and astrocytes.
Collapse
Affiliation(s)
- Benjamin Keller
- Laboratori de Neurofarmacologia, IUNICS-IdISPa, Universitat de les Illes Balears, Palma de Mallorca, Spain and Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| | - Jesús A García-Sevilla
- Laboratori de Neurofarmacologia, IUNICS-IdISPa, Universitat de les Illes Balears, Palma de Mallorca, Spain and Redes Temáticas de Investigación Cooperativa en Salud-Red de Trastornos Adictivos (RETICS-RTA), ISCIII, Madrid, Spain
| |
Collapse
|